WO2010043047A1 - Conjugates of glp-1 agonists and uses thereof - Google Patents

Conjugates of glp-1 agonists and uses thereof Download PDF

Info

Publication number
WO2010043047A1
WO2010043047A1 PCT/CA2009/001476 CA2009001476W WO2010043047A1 WO 2010043047 A1 WO2010043047 A1 WO 2010043047A1 CA 2009001476 W CA2009001476 W CA 2009001476W WO 2010043047 A1 WO2010043047 A1 WO 2010043047A1
Authority
WO
WIPO (PCT)
Prior art keywords
glp
compound
angiopep
ser
lys
Prior art date
Application number
PCT/CA2009/001476
Other languages
English (en)
French (fr)
Inventor
Jean-Paul Castaigne
Michel Demeule
Catherine Gagnon
Betty Lawrence
Original Assignee
Angiochem Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Angiochem Inc. filed Critical Angiochem Inc.
Priority to EP09820161.9A priority Critical patent/EP2346906A4/en
Priority to JP2011531313A priority patent/JP2012505637A/ja
Priority to BRPI0920209A priority patent/BRPI0920209A2/pt
Priority to AU2009304560A priority patent/AU2009304560A1/en
Priority to RU2011118056/10A priority patent/RU2011118056A/ru
Priority to CA2740316A priority patent/CA2740316A1/en
Priority to CN2009801502163A priority patent/CN102245642A/zh
Priority to US13/124,013 priority patent/US8921314B2/en
Priority to MX2011004017A priority patent/MX2011004017A/es
Publication of WO2010043047A1 publication Critical patent/WO2010043047A1/en
Priority to ZA2011/03044A priority patent/ZA201103044B/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/57563Vasoactive intestinal peptide [VIP]; Related peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif

Definitions

  • the invention relates to compounds including a GLP-I agonist (e.g., exendin-4), bound to a peptide vector and uses thereof.
  • GLP-I agonist e.g., exendin-4
  • uses include the treatment, prevention, and reduction of metabolic disorders including diabetes and obesity.
  • a GLP-I agonist e.g., exendin-4
  • Such uses include the treatment, prevention, and reduction of metabolic disorders including diabetes and obesity.
  • GLP-I agonist e.g., exendin-4
  • Such uses include the treatment, prevention, and reduction of metabolic disorders including diabetes and obesity.
  • Such uses include the treatment, prevention, and reduction of metabolic disorders including diabetes and obesity.
  • Loss of glucose homeostasis as a result of faulty insulin secretion or action typically results in metabolic disorders such as diabetes,
  • diabetes may arise secondary to any condition that causes extensive damage to the pancreas (e.g., pancreatitis, tumors, administration of certain drugs such as corticosteroids or pentamidine, iron overload (e.g., hemochromatosis), acquired or genetic endocrinopathies, and surgical excision), the most common forms of diabetes typically arise from primary disorders of the insulin signaling system.
  • type 1 diabetes also known as insulin dependent diabetes (IDDM)
  • type 2 diabetes also known as insulin independent or non-insulin dependent diabetes (NIDDM)
  • Type 1 diabetes which accounts for approximately 10% of all cases of primary diabetes, is an organ-specific autoimmune disease characterized by the extensive destruction of the insulin-producing beta cells of the pancreas. The consequent reduction in insulin production inevitably leads to the deregulation PATENT
  • ATTORNEY DOCKET NO. V82690WO of glucose metabolism While the administration of insulin provides significant benefits to patients suffering from this condition, the short serum half-life of insulin is a major impediment to the maintenance of normoglycemia.
  • An alternative treatment is islet transplantation, but this strategy has been associated with limited success.
  • Type 2 diabetes which affects a larger proportion of the population, is characterized by a deregulation in the secretion of insulin and/or a decreased response of peripheral tissues to insulin, i.e., insulin resistance. While the pathogenesis of type 2 diabetes remains unclear, epidemiologic studies suggest that this form of diabetes results from a collection of multiple genetic defects or polymorphisms, each contributing its own predisposing risks and modified by environmental factors, including excess weight, diet, inactivity, drugs, and excess alcohol consumption. Although various therapeutic treatments are available for the management of type 2 diabetes, they are associated with various debilitating side effects. Accordingly, patients diagnosed with or at risk of having type 2 diabetes are often advised to adopt a healthier lifestyle, including loss of weight, change in diet, exercise, and moderate alcohol intake.
  • a GLP-I agonist e.g., exendin-4
  • a peptide vector capable of transporting the GLP-I agonist either across the blood-brain barrier (BBB) or into a particular cell type (e.g., liver, lung, kidney, spleen, and muscle).
  • BBB blood-brain barrier
  • conjugates are targeted across the BBB or to particular cell types, therapeutic efficacy can be achieved using lower doses or less frequent dosings as compared to unconjugated GLP-I agonists, thus reducing the severity of or incidence of side effects and/or increasing efficacy.
  • the conjugate may also exhibit increased stability, improved pharmacokinetics, or reduced degradation in vivo.
  • the invention features a compound having the formula:
  • A-X-B where A is a peptide vector capable of being transported across the blood-brain barrier (BBB) or into a particular cell type (e.g., liver, lung, kidney, spleen, and muscle), X is a linker, and B is a GLP-I agonist (e.g., any described herein such as a peptide agonist).
  • BBB blood-brain barrier
  • B is a GLP-I agonist (e.g., any described herein such as a peptide agonist).
  • the transport across the BBB or into the cell may be increased by at least 10%, 25%, 50%, 75%, 100%, 200%, 500%, 750%, 1000%, 1500%, 2000%, 5000%, or 10,000%.
  • the compound may be substantially pure.
  • the compound may be formulated with a pharmaceutically acceptable carrier (e.g., any described herein).
  • the invention features methods of making the compound A-X-B.
  • the method includes conjugating the peptide vector (A) to a linker (X), and conjugating the peptide vector-linker (A- X) to a GLP-I agonist (B), thereby forming the compound A-X-B.
  • the method includes conjugating the GLP-I agonist (B) to a linker (X), and conjugating the GLP-I agonist/linker (X-B) to a peptide vector (A), thereby forming the compound A-X-B.
  • the method includes conjugating the peptide vector (A) to a GLP-I agonist (B), where either A or B optionally include a linker (X), to form the compound A-X-B.
  • the invention features a nucleic acid molecule that encodes the compound A-X-B, where the compound is a polypeptide.
  • the nucleic acid molecule may be operably linked to a promoter and may be part of a nucleic acid vector.
  • the vector may be in a cell, such as a prokaryotic cell PATENT
  • bacterial cell e.g., bacterial cell
  • eukaryotic cell e.g., yeast or mammalian cell, such as a human cell
  • the invention features methods of making a compound of the formula A-X-B, where A-X-B is a polypeptide.
  • the method includes expressing a nucleic acid vector of the previous aspect in a cell to produce the polypeptide; and purifying the polypeptide.
  • the invention features a method of treating (e.g., prophylactically) a subject having a metabolic disorder.
  • the method includes administering a compound of the first aspect in an amount sufficient to treat the disorder.
  • the metabolic disorder may be diabetes (e.g., Type I or Type II), obesity, diabetes as a consequence of obesity, hyperglycemia, dyslipidemia, hypertriglyceridemia, syndrome X, insulin resistance, impaired glucose tolerance (IGT), diabetic dyslipidemia, hyperlipidemia, a cardiovascular disease, or hypertension.
  • the invention features a method of reducing food intake by, or reducing body weight of, a subject. The method includes administering a compound of the first aspect to a subject in an amount sufficient to reduce food intake or reduce body weight.
  • the invention features a method of treating (e.g., prophylactically) a disorder selected from the group consisting of anxiety, movement disorder, aggression, psychosis, seizures, panic attacks, hysteria, sleep disorders, Alzheimer's disease, and Parkinson's disease.
  • the method includes administering a compound of the first aspect to a subject in an amount sufficient to treat or prevent the disorder.
  • the invention also features a method of increasing neurogenesis in a subject.
  • the method includes administering a compound of the first aspect to a subject.
  • the subject may desire, or may be in need of neurogenesis.
  • the subject may be suffering from a disease or disorder of the central nervous system such as Parkinson's Disease, Alzheimer's Disease, PATENT
  • the increase in neurogenesis can improve learning or enhance neuroprotection.
  • the invention features a method for converting liver stem/progenitor cells into functional pancreatic cells; preventing beta-cell deterioration and stimulation of beta-cell proliferation; treating obesity; suppressing appetite and inducing satiety; treating irritable bowel syndrome; reducing the morbidity and/or mortality associated with myocardial infarction and stroke; treating acute coronary syndrome characterized by an absence of Q- wave myocardial infarction; attenuating post-surgical catabolic changes; treating hibernating myocardium or diabetic cardiomyopathy; suppressing plasma blood levels of norepinepherine; increasing urinary sodium excretion, decreasing urinary potassium concentration; treating conditions or disorders associated with toxic hypervolemia, e.g., renal failure, congestive heart failure, nephrotic syndrome, cirrhosis, pulmonary edema, and hypertension; inducing an inotropic response and increasing cardiac contractility; treating polycystic ovary syndrome; treating respiratory distress; improving nutrition via a non- alimentary,
  • the invention features a method of increasing GLP-I receptor activity in a subject.
  • the method includes administering a compound of the first aspect to a subject in an amount sufficient to increase GLP-I receptor activity.
  • the method may reduce glucose levels in a subject.
  • the amount sufficient may be less than 90%, 75%, 50%, 40%, 30%, 20%, 15%, 10%, 5%, 4%, 3%, 2%, 1%, or 0.1% of the amount required for an equivalent dose of the GLP-I agonist when not conjugated to the peptide vector.
  • the amount sufficient may reduce side effects (e.g., vomiting, nausea, or diarrhea) as compared to administration of an effective amount of the GLP- 1 agonist when not conjugated to the peptide vector.
  • the subject may be a mammal such as a human.
  • the peptide vector may be a polypeptide substantially identical to any of the sequences set Table 1, or a fragment thereof.
  • the vector polypeptide has a sequence of
  • Angiopep-1 (SEQ ID NO:67), Angiopep-2 (SEQ ID NO:97), Angiopep-3 (SEQ ID NO:107), Angiopep-4a (SEQ ID NO:108), Angiopep-4b (SEQ ID NO:109), Angiopep-5 (SEQ ID NO:110), Angiopep-6 (SEQ ID NO: 111), or Angiopep-7 (SEQ ID NO: 112)).
  • the peptide vector or conjugate may be efficiently transported into a particular cell type (e.g., any one, two, three, four, or five of liver, lung, kidney, spleen, and muscle) or may cross the mammalian BBB efficiently (e.g., Angiopep-1, -2, -3, -4a, -4b, -5, and -6).
  • a particular cell type e.g., any one, two, three, four, or five of liver, lung, kidney, spleen, and muscle
  • the peptide vector may be of any length, for example, at least 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 25, 35, 50, 75, 100, 200, or 500 amino acids, or any range between these numbers. In certain embodiments, the peptide vector is 10 to 50 amino acids in length.
  • the polypeptide may be produced by recombinant genetic technology or chemical synthesis.
  • Polypeptides Nos.5, 67, 76, and 91 include the sequences of SEQ ID NOS:5, 67, 76, and 91, respectively, and are amidated at the C-terminus.
  • Polypeptides Nos.107, 109, and 110 include the sequences of SEQ ID NOS:97, 109,and 110, respectively, and are acetylated at the N-terminus.
  • the peptide vector may include an amino acid sequence having the formula:
  • X1-X19 e.g., X1-X6, X8, X9, X11-X14, and X16-X19
  • X1-X19 is, independently, any amino acid (e.g., a naturally occurring amino acid such as Ala, Arg, Asn, Asp, Cys, GIn, GIu, GIy, His, He, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, and VaI) or absent and at least one (e.g., 2 or 3) of Xl, XlO, and X15 is arginine.
  • a naturally occurring amino acid such as Ala, Arg, Asn, Asp, Cys, GIn, GIu, GI
  • X7 is Ser or Cys; or XlO and X15 each are independently Arg or Lys.
  • the residues from Xl through X 19, inclusive are substantially identical to any of the amino acid sequences of any one of SEQ ID NOS: 1-105 and 107-116 (e.g., Angiopep-1, Angiopep-2, Angiopep-3, Angiopep-4a, Angiopep-4b, Angiopep-5, Angiopep- PATENT
  • the GLP- 1 agonist may be a peptide agonist.
  • the GLP-I agonist may GLP-I, exendin-4, exendin-3, or analog or fragment thereof (e.g., any analog or fragment described herein).
  • the GLP-I agonist is an exendin-4 analog selected from the group consisting of [Lys 39 ]exendin-4 and [Cys 32 ]exendin-4.
  • the peptide vector or peptide GLP-I agonist is modified (e.g., as described herein).
  • the polypeptide may be amidated, acetylated, or both. Such modifications to polypeptides may be at the amino or carboxy terminus of the polypeptide.
  • the polypeptide may also include peptidomimetics (e.g., those described herein) of any of the polypeptides described herein.
  • the polypeptide may be in a multimeric form, for example, dimeric form (e.g., formed by disulfide bonding through cysteine residues).
  • the polypeptide has an amino acid sequence described herein with at least one amino acid substitution (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 substitutions), insertion, or deletion.
  • the polypeptide may contain, for example, 1 to 12, 1 to 10, 1 to 5, or 1 to 3 amino acid substitutions, for example, 1 to 10 (e.g., to 9, 8, 7, 6, 5, 4, 3, 2) amino acid substitutions.
  • the amino acid substitution(s) may be conservative or non-conservative.
  • the peptide vector may have an arginine at one, two, or three of the positions corresponding to positions 1, 10, and 15 of the amino acid sequence of any of SEQ ID NO:1, Angiopep-1, Angiopep-2, Angiopep-3, Angiopep-4a, Angiopep-4b, Angiopep-5, Angiopep-6, and Angiopep-7.
  • the GLP-I agonist may have a cysteine or lysine substitution or addition at any position (e.g., a lysine substitution at the N- or C-terminal position, or a cysteine substitution at the position corresponding to amino acid 32 of the exendin-4 sequence).
  • the compound may specifically exclude a polypeptide including or consisting of any of SEQ ID NOS: 1-105 and 107-116 (e.g., Angiopep-1, Angiopep-2, Angiopep-3, Angiopep-4a, Angiopep-4b, Angiopep-5, Angiopep-6, and Angiopep-7).
  • the polypeptides and conjugates of the invention exclude the polypeptides of SEQ ID NOs: 102, 103, 104, and 105.
  • the linker (X) may be any linker known in the art or described herein.
  • the linker is a covalent bond (e.g., a peptide bond), a chemical linking agent (e.g., those described herein), an amino acid or a peptide (e.g., 2, 3, 4, 5, 8, 10, or more amino acids).
  • the linker has the formula:
  • n is an integer between 2 and 15 (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15); and either Y is a thiol on A and Z is a primary amine on B or Y is a thiol on B and Z is a primary amino on A.
  • GLP-I agonist any compound capable of activating a GLP-I receptor (e.g., a mammalian or human GLP-I receptor).
  • Agonists can include peptides or small molecule compounds (e.g., any of those described herein).
  • Assays for determining whether a particular compound is a GLP-I agonist are known in the art and described herein.
  • peptide vector is meant a compound or molecule such as a polypeptide or a polypeptide mimetic that can be transported into a particular cell type (e.g., liver, lungs, kidney, spleen, or muscle) or across the BBB.
  • the vector may be attached to (covalently or not) or conjugated to an agent (e.g., a GLP- 1 agonist) and thereby may be able to transport the agent into a particular cell type or across the BBB.
  • the vector may bind to receptors present on cancer cells or brain endothelial cells and thereby be PATENT
  • the vector may be a molecule for which high levels of transendothelial transport may be obtained, without affecting the cell or BBB integrity.
  • the vector may be a polypeptide or a peptidomimetic and may be naturally occurring or produced by chemical synthesis or recombinant genetic technology.
  • substantially identical is meant a polypeptide or nucleic acid exhibiting at least 35%, 40%, 50%, 55%, 60%, 65%, 70%, 75%, 85%, 90%, 95%, or even 99% identity to a reference amino acid or nucleic acid sequence.
  • the length of comparison sequences will generally be at least 4 (e.g., at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 50, or 100) amino acids.
  • the length of comparison sequences will generally be at least 60 nucleotides, preferably at least 90 nucleotides, and more preferably at least 120 nucleotides, or full length.
  • gaps may be found between the amino acids of an analogs that are identical or similar to amino acids of the original polypeptide.
  • the gaps may include no amino acids, one or more amino acids that are not identical or similar to the original polypeptide.
  • Biologically active analogs of the vectors (polypeptides) of the invention are encompassed herewith. Percent identity may be determined, for example, with n algorithm GAP, BESTFIT, or FASTA in the Wisconsin Genetics Software Package Release 7.0, using default gap weights.
  • treating a disease, disorder, or condition in a subject is meant reducing at least one symptom of the disease, disorder, or condition by administrating a therapeutic agent to the subject.
  • treating prophylactically a disease, disorder, or condition in a subject is meant reducing the frequency of occurrence of (e.g., preventing) a disease, disorder or condition or reducing the severity of the disease, disorder, or condition by administering a therapeutic agent to the subject.
  • a subject who is being treated for a metabolic disorder is one who a medical practitioner has diagnosed as having such a condition. Diagnosis may be performed by any suitable means, such as those described herein.
  • ATTORNEY DOCKET NO. V82690WO in whom the development of diabetes or obesity is being prevented may or may not have received such a diagnosis.
  • subject of the invention may have been subjected to standard tests or may have been identified, without examination, as one at high risk due to the presence of one or more risk factors, such as family history, obesity, particular ethnicity (e.g., African Americans and Hispanic Americans), gestational diabetes or delivering a baby that weighs more than nine pounds, hypertension, having a pathological condition predisposing to obesity or diabetes, high blood levels of triglycerides, high blood levels of cholesterol, presence of molecular markers (e.g., presence of autoantibodies), and age (over 45 years of age).
  • risk factors such as family history, obesity, particular ethnicity (e.g., African Americans and Hispanic Americans), gestational diabetes or delivering a baby that weighs more than nine pounds, hypertension, having a pathological condition predisposing to obesity or diabetes, high blood levels of trig
  • a metabolic disorder is meant any pathological condition resulting from an alteration in a subject's metabolism. Such disorders include those resulting from an alteration in glucose homeostasis resulting, for example, in hyperglycemia. According to this invention, an alteration in glucose levels is typically an increase in glucose levels by at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or even 100% relative to such levels in a healthy individual. Metabolic disorders include obesity and diabetes (e.g., diabetes type I, diabetes type II, MODY, and gestational diabetes), satiety, and endocrine deficiencies of aging.
  • reducing glucose levels is meant reducing the level of glucose by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100% relative to an untreated control.
  • glucose levels are reduced to normoglycemic levels, i.e., between 150 to 60 mg/dL, between 140 to 70 mg/dL, between 130 to 70 mg/dL, between 125 to 80 mg/dL, and preferably between 120 to 80 mg/dL.
  • normoglycemic levels i.e., between 150 to 60 mg/dL, between 140 to 70 mg/dL, between 130 to 70 mg/dL, between 125 to 80 mg/dL, and preferably between 120 to 80 mg/dL.
  • Such reduction in glucose levels may be obtained by PATENT
  • ATTORNEY DOCKET NO. V82690WO increasing any one of the biological activities associated with the clearance of glucose from the blood (e.g., increase insulin production, secretion, or action).
  • subject is meant a human or non-human animal (e.g., a mammal).
  • increasing GLP-I receptor activity is meant increasing the level of receptor activation measured using standard techniques (e.g., cAMP activation) by, for example, at least %, 20%, 50%, 75%, 100%, 200%, or 500% as compared to an untreated control.
  • equivalent dosage is meant the amount of a compound of the invention required to achieve the same molar amount of the GLP-I agonist in the compound, as compared to the unconjugated GLP-I agonist.
  • equivalent dosage of 1.0 ⁇ g exendin-4 is about 1.6 ⁇ g of the [Lys 39 - MHA]exendin-4/Angiopep-2-Cys-NH 2 conjugate described herein.
  • a polypeptide which is "efficiently transported across the BBB” is meant a polypeptide that is able to cross the BBB at least as efficiently as Angiopep-6 (i.e., greater than 38.5% that of Angiopep- 1 (250 nM) in the in situ brain perfusion assay described in U.S. Patent Application No. 11/807,597, filed May 29, 2007, hereby incorporated by reference). Accordingly, a polypeptide which is "not efficiently transported across the BBB” is transported to the brain at lower levels (e.g., transported less efficiently than Angiopep-6).
  • polypeptide or compound which is "efficiently transported to a particular cell type” is meant that the polypeptide or compound is able to accumulate (e.g., either due to increased transport into the cell, decreased efflux from the cell, or a combination thereof) in that cell type to at least a 10% (e.g., 25%, 50%, 100%, 200%, 500%, 1,000%, 5,000%, or 10,000%) greater extent than either a control substance, or, in the case of a conjugate, as compared to the unconjugated agent.
  • a 10% e.g., 25%, 50%, 100%, 200%, 500%, 1,000%, 5,000%, or 10,000% greater extent than either a control substance, or, in the case of a conjugate, as compared to the unconjugated agent.
  • Figure 1 is table and schematic diagram showing exendin-4 and the exendin-4 analogs used in experiments described herein.
  • Figure 2 is a schematic diagram of the synthetic scheme used to conjugate Cys-AngioPep2, Angiopep-2-Cys-NH 2 , and Angiopep-1 to [Lys 39 - MHA]exendin-4.
  • Figure 3 is a schematic diagram of the synthetic scheme used to conjugate [Cys 32 ] exendin-4 to (maleimido propionic acid (MPA))-Angiopep-2, (maleimido hexamoic acid (MHA))- Angiopep-2, and (maleimido undecanoic acid (MU A))- Angiopep-2.
  • MPA maleimido propionic acid
  • MHA maleimido hexamoic acid
  • MU A maleimido undecanoic acid
  • Figure 4 is a graph showing transport of exendin-4 (left) and exendin- 4/Angiopep-2 (N- terminal, center; c-terminal, right) across the BBB. The total amount in the brain, along with the amounts in the capillaries and the parenchyma are shown.
  • Figure 5 is a graph showing increase in weight of (ob/ob) mice following administration of a control, exendin-4, or the [Lys 39 -MHA]exendin- 4/Angiopep-2-Cys-NH 2 conjugate (Exen-An2). Both exendin-4 and Ex- An2 were observed to reduce weight gain as compared to the animals receiving the control.
  • Figure 6 is a graph showing total food consumption by (ob/ob) mice, where the mice were administered a control, exendin-4, or the Exen-An2. Both exendin-4 and Exen-An2 were observed to reduce food intake as compared to the animals receiving the control.
  • Figure 7 is a graph showing reduction in glycemia following administration of two doses of exendin-4 (3 ⁇ g/kg and 30 ⁇ g/kg) and equivalent doses of Exen-An2 (4.8 ⁇ g/kg and 48 ⁇ g/kg). A similar reduction in glycemia at the lower dose of Exen-An2, as compared to the higher dose of exendin-4, was observed. During this experiment, one mouse in the control group died at day 12.
  • Figure 8A is a schematic diagram showing the structure of an Exendin- 4-Angiopep-2 dimer conjugate (Ex4(Lys39(MHA))-AN2-AN2).
  • the compound has the structure
  • Figure 8B is a schematic structure of an Exendin-4-scramble-Angiopep- 2 (Ex4(Cys32)-ANS4 (N-Term) or Exen-S4) that was used a control. This compound has the structure HGEGTFTSDLSKQMEEEAVRLFIEWLKNGGPCSGAPPPS-(MHA)-
  • FIG. 9 is a graph showing the ability of, from left to right, Exendin-4; Exendin-4-Angiopep-2 conjugates C3, C6, and CI l; Exen-S4; and Exendin-4 when conjugated to a dimeric form of Angiopep-2, to cross the BBB.
  • Figure 10 is a graph showing the ability of Exendin-4 and Exen-An2-
  • mice An2 to reduce glycemia in mice.
  • GLP-I agonist/peptide conjugates having an enhanced ability to cross the blood-brain barrier (BBB) or to enter particular cell type(s) (e.g., liver, lung, kidney, spleen, and muscle) using the exemplary GLP-I agonist exendin-4 and exendin-4 analogs.
  • the peptide conjugates of the invention can include a GLP-I agonist and a peptide vector that enhance transport across the BBB.
  • lower doses of the compounds of the invention, as compared to unconjugated GLP-I agonists are effective in treating GLP-I related disorders including a reduction in glycemia.
  • side effects such as vomiting, nausea, and diarrhea observed with the unconjugated agonists can be reduced or eliminated.
  • increased efficacy at higher doses may be obtained.
  • the GLP-I agonist can be any GLP-I agonist known in the art and including peptides such as those described below.
  • Particular GLP-I agonists include exendin-4, GLP-I, and exendin-3 fragments, substitutions (e.g., conservative or nonconservative substitutions, or substitutions of non-naturally occurring amino acids), and chemical modifications to the amino acid sequences (e.g., those described herein).
  • Particular GLP-I agonists are described in detail below.
  • GLP-I agonists The conjugates of the invention can include any GLP- 1 agonist known in the art.
  • Particular GLP-I agonists include GLP-I, exendin-4, and analogs thereof. Exemplary analogs are described below.
  • Exendin-4 and exendin-4 analogs can also be used in the compositions, methods, and kits of the invention.
  • the compounds of the invention can include fragments of the exendin-4 sequence.
  • Exendin-4 has the sequence.
  • exendin-4 analogs include those having a cysteine substitution (e.g., [Cys 32 ] exendin-4) or a lysine substitution (e.g., [Lys 39 ] exendin-4).
  • Exendin analogs are also described in U.S. Patent No. 7,157,555 and include those of the formula:
  • X 5 is Thr or Ser
  • X 6 is Ser or Thr
  • X 7 is Asp or GIu
  • X 8 is Leu, Ue, VaI, pGly or Met
  • X 9 is Leu, He, pGly, VaI or Met
  • X 10 is Phe, Tyr, or NaI
  • X 11 is He, VaI, Leu, pGly, t-BuG or Met
  • X 12 is GIu or Asp
  • X 13 is Trp, Phe, Tyr, or NaI
  • X 14 , X 15 , X 16 and X 17 are independently Pro, HPro, 3Hyp, 4Hyp, TPro, N-alkylglycine, N-alkyl-pGly, or N-alkylalanine
  • X 18 is Ser, Thr, or Tyr
  • Z is -OH or -NH 2 (e.g., with the proviso that the compound is not exendin- 3 or exindin-4.)
  • N-alkyl groups for N-alkylglycine, N-alkyl-pGly and N- alkylalanine include lower alkyl groups (e.g., Ci -6 alkyl or Ci ⁇ alkyl).
  • X 1 is His or Tyr (e.g., His).
  • X 2 can be GIy.
  • X 9 can be Leu, pGly, or Met.
  • Xj 3 can be Trp or Phe.
  • X 4 can be Phe or NaI;
  • X 11 can be He or VaI, and
  • Xi 4 , X ]5 , X )6 and X] 7 can be independently selected from Pro, HPro, TPro, or N-alkylalanine (e.g., where N-alkylalanine has a N-alkyl group of 1 to about 6 carbon atoms).
  • X 15 , X 16 , and X 17 are the same amino acid residue.
  • X )8 may be Ser or Tyr (e.g., Ser).
  • Z can be -NH 2 .
  • X) is His or Tyr (e.g., His);
  • X 2 is GIy;
  • X 4 is Phe or NaI;
  • X 9 is Leu, pGly, or Met;
  • Xi 0 is Phe or NaI;
  • Xj ] is He or VaI;
  • X 14 , X 15 , X 16 , and X 17 are independently selected from Pro, HPro, TPro, or N- alkylalanine; and
  • Xj 8 is Ser or Tyr, (e.g., Ser).
  • Z can be -NH 2 .
  • X] is His or Arg; X 2 is GIy; X 3 is Asp or GIu; X 4 is Phe or napthylalanine; X 5 is Thr or Ser; X 6 is Ser or Thr; X 7 is Asp or GIu; Xg is Leu or pGly; X 9 is Leu or pGly; Xj 0 is Phe or NaI; Xi ] is He, VaI, or t- butyltylglycine; X ]2 is GIu or Asp; Xn is Trp or Phe; X )4 , X ]5 , X ]6 , and X ]7 are independently Pro, HPro, TPro, or N-methylalanine; X] 8 is Ser or Tyr: and Z is -OH Or-NH 2 (e.g., where the compound is not exendin-3 or exendin-4).
  • Z can be -NH 2 .
  • X 9 is Leu, He, VaI, or pGly (e.g., Leu or pGly) and Xi 3 is Phe, Tyr, or NaI (e.g., Phe or NaI).
  • Xi 3 is Phe, Tyr, or NaI (e.g., Phe or NaI).
  • Xi is His, Arg, or Tyr
  • X 2 is Ser, GIy, Ala, or Thr
  • X 3 is Asp or GIu
  • X 5 is Ala or Thr
  • X 6 is Ala, Phe, Tyr, or NaI
  • X 7 is Thr or Ser
  • X 8 is Ala, Ser, or Thr
  • X 9 is Asp or GIu
  • X ]0 Ala, Leu, He, VaI, pGly, or Met
  • X 11 is Ala or Ser
  • N-alkyl groups for N-alkylglycine, N-alkyl-pGly and N- alkylalanine include lower alkyl groups of 1 to about 6 carbon atoms (e.g., 1 to
  • X 1 is His or Tyr (e.g., His).
  • X 2 can be GIy.
  • Xi 4 can be Leu, pGly, or Met.
  • X 25 can be Trp or Phe.
  • X 6 is Phe or NaI
  • X 22 is Phe or NaI
  • X 23 is He or VaI.
  • X 3f , X 36 , X 37 , and X 38 can be independently selected from Pro, HPro, TPro, and N-alkylalanine.
  • Z 1 is -NH 2 or Z 2 is -NH 2 .
  • X) is His or Tyr (e.g., His);
  • X 2 is GIy;
  • X 6 is Phe or NaI;
  • X 14 is Leu, pGly, or Met;
  • X 22 is Phe or NaI;
  • X 23 is He or VaI;
  • X 31 , X 36 , X 37 , and X 38 are independently selected from Pro, HPro, TPro, or N- alkylalanine.
  • Z 1 is -NH 2 .
  • X 1 is His or Arg;
  • X 2 is GIy or Ala;
  • X 3 is Asp or GIu;
  • X 5 is Ala or Thr;
  • X 6 is Ala, Phe, or naphthylalanine;
  • X 7 is Thr or Ser;
  • X g is Ala, Ser, or Thr;
  • X 9 is Asp or GIu;
  • X] 0 Ala, Leu, or pGly;
  • Xn is Ala or Ser;
  • Xi 2 is Ala or Lys;
  • X ]3 is Ala or GIn;
  • X )4 is Ala, Leu, or pGly;
  • X 15 is Ala or GIu;
  • X 16 is Ala or GIu;
  • X 17 is Ala or GIu;
  • X 19 is Ala or VaI;
  • X 20 is Ala or Arg;
  • X 21 is
  • Xl2 Xl 3 , Xl4j XiS, Xl6> Xl7> Xl9, X20, X2U ⁇ 24, X25> ⁇ 26? ⁇ 27 an ⁇ X28 ⁇ S Ala).
  • Xi 4 is Leu, lie, VaI, or pGly (e.g., Leu or pGly), and X 25 is Phe, Tyr or NaI (e.g., Phe or NaI).
  • Exendin analogs described in U.S. Patent No. 7,220,721 include compounds of the formula:
  • X21-X 22 -X23-X24-X25-X26-X27-X28-Zi where X 1 is His, Arg, Tyr, Ala, Norval, VaI, or Norleu; X 2 is Ser, GIy, Ala, or Thr; X 3 is Ala, Asp, or GIu; X 4 is Ala, Norval, VaI, Norleu, or GIy; X 5 is Ala or Thr; X 6 is Phe, Tyr or NaI; X 7 is Thr or Ser; X 8 is Ala, Ser or Thr; X 9 is Ala, Norval, VaI, Norleu, Asp, or GIu; Xj 0 is Ala, Leu, He, VaI, pGly, or Met; Xn is Ala or Ser; X 12 is Ala or Lys; X ⁇ 3 is Ala or GIn; X H is Ala, Leu, He, pGly, VaI, or Met
  • X ⁇ > Xi2j Xn, Xi4, Xi5 > Xi6> X ⁇ > Xi9> ⁇ 2O 5 X21, X24, ⁇ 25» X26, X27 and X 2 g are Ala and/or provided also that, if X 1 is His, Arg, or Tyr, then at least one of X 3 , X 4 and X 9 is Ala).
  • exendin-4 analogs include exendin-4(l-30), exendin-4(l-30) amide, exendin-4(l-28) amide, [Leu 14 ,Phe 25 ]exendin-4 amide, [Leu 14 ,Phe 25 ]exendin-4(l-28) amide, and [Leu 14 ,Ala 22 ,Phe 25 ]exendin-4(l-28) amide.
  • U.S. Patent No. 7,329,646 describes exendin-4 analogs having the general formula:
  • exendin-4 derivatives include [(Ile/Leu/Met) 14 ,(His/Lys) 20 ,Arg 40 ]exendin-4; [(not Lys/not Arg) 12 ,(not Lys/not Arg) 20 ,(not Lys/not Arg) 27 ,Arg 40 ]exendin-4; and [(not Lys/not Arg) 20 ,Arg 40 ]exendin-4.
  • exendin-4 analogs include [Lys 20 ,Arg 4 °]exendin-4,[His 20 ,Arg 40 ]exendin-4; and [Leu 14 ,Lys 20 ,Arg 40 ]exendin-4.
  • the invention may also use truncated forms of exendin-4 or any of the exendin analogs described herein.
  • the truncated forms may include deletions of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino PATENT
  • ATTORNEY DOCKET NO. V82690WO acids from the N-terminus, from the C-terminus, or a combination thereof.
  • exendin-4 fragments include Exendin-4(1-31).
  • Other fragments of exendin-4 are described in U.S. Patent Application Publication No. 2007/0037747 and have the formula:
  • the GLP-I agonist used in the compositions, methods, and kits of the invention can be GLP- 1 or a GLP- 1 analog.
  • the GLP- 1 analog is a peptide, which can be truncated, may have one or more substitutions of the wild type sequence (e.g., the human wild type sequence), or may have other chemical modifications.
  • GLP-I agonists can also be non- peptide compounds, for example, as described in U.S. Patent No. 6,927,214. Particular analogs include LY548806, CJC-1131, and Liraglutide.
  • the GLP- 1 analog can be truncated form of GLP- 1.
  • the GLP- 1 peptide may be truncated by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 15, 20, or more residues from its N-terminus, its C-terminus, or a combination thereof.
  • the truncated GLP-I analog is the GLP- 1(7-34), GLP- 1(7-35), GLP-l(7-36), or GLP-I (7-37) human peptide or the C-terminal amidated forms thereof.
  • modified forms of truncated GLP-I peptides are used.
  • Exemplary analogs are described in U.S. Patent No. 5,545,618 and have the amino acid sequence:
  • the substituted amino acids may be in the D form.
  • the amino acids substituted at position 7 can also be the N-acylated or N-alkylated amino acids.
  • Exemplary GLP-I analogs include [D-His 7 ]GLP- 1(7-37), [Tyr 7 ]GLP-l(7-37), [N-acetyl-His 7 ]GLP- 1(7-37), [N-isopropyl- His 7 ]GLP- 1(7-37), [D-AIa 8 JGLP- 1(7-37), [D-GIu 9 ] GLP- 1(7-37), [Asp 9 ]GLP- 1(7-37), [D-Asp 9 ]GLP- 1(7-37), [D-Asp 9 ]GLP- 1(7-37), [D-Phe l0 ]GLP- 1(7-37),
  • Gly-Arg-R ⁇ 2 where R, is H 2 N; H 2 N-Ser; H 2 N-Val-Ser; H 2 N-Asp-Val-Ser; H 2 N-Ser-Asp-Val- Ser; H 2 N-Thr-Ser-Asp-Val-Ser; H 2 N-Phe-Thr-Ser-Asp-Val-Ser; H 2 N-Thr-Phe- Thr-Ser-Asp-Val-Ser; H 2 N-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser; H 2 N-GIu-GIy- PATENT
  • X is Lys or Arg; and
  • R 2 is NH 2 , OH, GIy-NH 2 , or GIy-OH.
  • GLP-I analogs described in U.S. Patent No. 5,118,666, include the sequence His-Ala-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu- Glu-Gly-Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-X, where X is Lys, Lys-Gly, or Lys-Gly-Arg.
  • GLP-I analogs also include peptides of the formula: H 2 N-X-CO-R 1 , where R 1 is OH, OM, or -NR 2 R 3 ; M is a pharmaceutically acceptable cation or a lower branched or unbranched alkyl group (e.g., C 1 6 alkyl); R 2 and R 3 are independently selected from the group consisting of hydrogen and a lower branched or unbranched alkyl group (e.g., Ci -6 alkyl); X is a peptide comprising the sequence His-Ala-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu- Glu-Gly-Gln- Ala-Ala -Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Lys-Gly-Arg; NH 2 is the amine group of the amino terminus of X; and CO is the carbonyl group
  • Ri is His, D-His, desamino-His, 2-amino-His, ⁇ -hydroxy-His, homohistidine, ⁇ -fluoromethyl-His, or ⁇ -methyl-His
  • X is Met, Asp, Lys, Thr, Leu, Asn, GIn, Phe, VaI, or Tyr
  • Y and Z are independently selected from GIu, GIn, Ala, Thr, Ser, and GIy
  • R 2 is selected from NH 2 and GIy-OH (e.g., provided that, AfR 1 is His, X is VaI, Y is GIu, and Z
  • Ri is 4-imidazopropionyl (des-amino-histidyl), 4-imidazoacetyl, or 4- imidazo- ⁇ , ⁇ dimethyl-acetyl
  • R 2 which is bound to the side chain of the Lys (e.g., through the ⁇ amino group), is C 6-10 unbranched acyl or is absent
  • R 3 is GIy-OH or NH 2
  • Xaa is Lys or Arg.
  • GLP-I analog has the formula:
  • X 8 is GIy, Ala, VaI, Leu, He, Ser, or Thr
  • X 11 is Asp, GIu, Arg, Thr, Ala, Lys, or His
  • X 12 is His, Trp, Phe, or Tyr
  • Xi 6 is Leu, Ser, Thr, Trp, His, Phe, Asp, VaI, Tyr, GIu, or Ala
  • X 22 is GIy, Asp, GIu, GIn, Asn, Lys, Arg, Cys, or Cya
  • X 23 is His, Asp, Lys
  • polypeptide has the amino acid sequence:
  • X 8 is GIy, Ala, VaI, Leu, He, Ser, or Thr
  • X 12 is His, Trp, Phe, or Tyr
  • X 16 is Leu, Ser, Thr, Trp, His, Phe, Asp, VaI, GIu, or Ala
  • X 22 is GIy, Asp, GIu, GIn, Asn, Lys, Arg, Cys, or Cya
  • X 23 is His, Asp, Lys, GIu, or GIn
  • X 26 is Asp, Lys, GIu, or His
  • X 30 is Ala, GIu, Asp,
  • polypeptide has the amino acid sequence:
  • X 8 is GIy, Ala, VaI, Leu, He, Ser, or Thr
  • X 22 is GIy, Asp, GIu, GIn, Asn, Lys, Arg, Cys, or Cya
  • X 23 is His, Asp, Lys, GIu, or GIn
  • X 27 is Ala, GIu, His, Phe, Tyr, Trp, Arg, or Lys
  • X 30 is Ala, GIu, Asp, Ser, or His
  • R is Lys, Arg, Thr, Ser, GIu, Asp, Trp, Tyr, Phe, His, -NH 2 , GIy
  • polypeptide has the amino acid sequence:
  • X 7 is L-His, D-His, desamino-His, 2amino-His, ⁇ -hydroxy-His, homo- His, ⁇ -fluoromethyl-His or ⁇ -methyl-His
  • X 8 is GIy, Ala, VaI, Leu, He, Ser or Thr (e.g., GIy, VaI, Leu, He, Ser, or Thr)
  • X 22 is Asp, GIu, GIn, Asn, Lys, Arg, Cys, or Cya
  • R is -NH 2 or GIy(OH).
  • the GLP-I compound has an amino acid other than alanine at position 8 and an amino acid other than glycine at position 22.
  • GLP-I compounds include [GIU 22 JGLP- I (7-37)OH, [Asp 22 ]GLP- 1 (7-37)OH, [Arg 22 ]GLP- 1 (7-37)OH, [Lys 22 ]GLP- 1 (7-37)OH, [Cya 22 ]GLP- 1 (7-37)OH, [Val 8 ,Glu 22 ]GLP- 1 (7-37)OH, [Val 8 ,Asp 22 ]GLP- 1 (7- 37)OH, [Val 8 ,Arg 22 ]GLP-l(7-37)OH, [Val 8 ,Lys 22 ] GLP-I (7-37)OH, [Val 8 ,Cya 22 ]GLP- 1 (7-37)OH, [Gly 8 ,Glu 22 ]GLP- 1 (7-37)OH, [Gly 8 ,
  • X 7 -X 8 -Glu-Gly-Thr-Xi 2 -Thr-Ser-Asp-X 16 -Ser-X 18 -X 19 -X 2O -Glu-X 22 -Gln-Ala- X 25 -Lys-X 27 -Phe-Ile-X 3 o-T ⁇ -Leu-X 33 -Lys-Gly-Arg-X 37
  • X 7 is L-His, D-His, desamino-His, 2-amino-His, ⁇ -hydroxy-His, homohistidine, ⁇ -fluoromethyl-His, or ⁇ -methyl-His
  • X 8 is Ala, GIy, VaI, Leu, He, Ser, or Thr
  • X 12 is Phe, Tip, or Tyr
  • X 16 is VaI, Trp, lie, Leu, Phe, or Tyr
  • X 18 is Ser, Trp, Tyr, Phe, Lys, He
  • ATTORNEY DOCKET NO. V82690WO substituted with one or more of H, alkyl, cycloalkyl, arylalkyl, aryl, heterocyclyl, heteroaryl, alkenyl, alkynyl, halo, hydroxy, mercapto, nitro, cyano, amino, acylamino, azido, guanidino, amidino, carboxyl, carboxamido, carboxamido alkyl, formyl, acyl, carboxyl alkyl, alkoxy, aryloxy, arylalkyloxy, heteroaryloxy, heterocycleoxy, acyloxy, mercapto, mercapto alkyl, mercaptoaryl, mercapto acyl, halo, cyano, nitro, azido, amino, guanidino alkyl, guanidino acyl, sulfonic, sulfonamido, alkyl s
  • Exemplary substitutions on the ⁇ -carbon atoms of Y and Z include heteroarylarylmethyl, arylheteroarylmethyl, and biphenylmethyl forming biphenylalanine residues, any of which is also optionally substituted with one or more, hydrogen, alkyl, cycloalkyl, arylalkyl, aryl, heterocyclyl, heteroaryl, alkenyl, alkynyl, halo, hydroxy, mercapto, nitro, cyano, amino, acylamino, azido, guanidino, amidino, carboxyl, carboxamido, carboxamido alkyl, fo ⁇ nyl, acyl, carboxyl alkyl, alkoxy, aryloxy, arylalkyloxy, heteroaryloxy, heterocycleoxy, acyloxy, mercapto, mercapto alkyl, mercaptoaryl, mercapto acyl, halo, cyano,
  • inventions include isolated polypeptides where the other substitution at the ⁇ -carbon of Y is substituted with H, methyl, or ethyl; and where the other substitution at the ⁇ -carbon of Z is substituted with H, methyl, or ethyl.
  • X 1 is naturally or non-naturally occurring amino acid residue in which one of the substitutions at the ⁇ -carbon is a primary substituent selected from the group consisting of heterocyclylalkyl, heteroaryl, heteroarylkalkyl and arylalkyl, said primary substituent optionally being substituted with secondary substituent selected from heteroaryl or heterocyclyl; and in which the other substitution at the ⁇ -carbon is H or alkyl;
  • X 2 is naturally or nonnaturally occurring amino acid residue in which one of the substitutions at the ⁇ -carbon is an alkyl or cycloalkyl where the alkyl group may optionally form a ring with the nitrogen of X 2 ; and wherein the other substitution at the ⁇ -carbon is H or alkyl;
  • X 3 is a naturally or nonnaturally occurring amino acid residue in which one of the substitutions at the ⁇ -carbon is a carboxyalkyl, bis-carboxyalkyl, sulfonyl
  • X 5 is a naturally or nonnaturally occurring amino acid residue in which one of the substitutions at the ⁇ -carbon is an alkyl or hydroxyalkyl, and in which the other substitution at the ⁇ -carbon is hydrogen or alkyl;
  • X 6 is a naturally or nonnaturally occurring amino acid residue in which one of the substitutions at the ⁇ -carbon is C] -12 alkyl, aryl, heteroaryl, heterocyclyl, cycloalkylalkyl, heterocyclylalkyl, arylalkyl, or heteroarylalkyl group, and the other substitution at the ⁇ -carbon is H or alkyl;
  • X 7 is a naturally or nonnaturally occurring amino acid residue in which
  • X 1 is His, D-His, N-Methyl-His, D-N-Methyl- His, 4-ThiazolylAla, or D-4-ThiazolylAla
  • X 2 is Ala, D-AIa, Pro, GIy, D-Ser, D-Asn, Nma, D-Nma, 4-ThioPro, 4-Hyp, L-2-Pip, L-2-Azt, Aib, S- or R-Iva and Acc3
  • X 3 is GIu, N-Methyl-Glu, Asp, D- Asp, His, GIa, Adp, Cys, or 4- ThiazolyAla
  • X 4 is GIy, His, Lys, or Asp
  • X 5 is Thr, D-Thr, NIe, Met, Nva, or L-Aoc
  • X 6 is Phe, Tyr, Tyr(Bzl), Tyr(3-NO 2 ), NIe,
  • Additional embodiments include those where Y is Bip, D-Bip, L-Bip(2- Me), D-Bip(2-Me), L-Bip(2'-Me), L-Bip(2-Et), D-Bip(2-Et), L-Bip(3-Et), L- Bip(4-Et), L-Bip(2-n- ⁇ ro ⁇ yl), L-Bi ⁇ (2-n-propyl, 4-OMe), L-Bip(2-n-pro ⁇ yl,2 '- Me), L-Bip(3-Me), L-Bip(4-Me), L-Bip(2,3-di-Me), L-Bip(2,4-di-Me), L- Bip(2,6-di-Me), L-Bip(2,4-di-Et), L-Bip(2-Me, 2'-Me), L-Bip(2-Et, 2'-M
  • Xj is an R group, an R-C(O) (amide) group, a carbamate group RO-C(O), a urea R 4 R 5 N-C(O), a sulfonamido R-SO 2 , or a R 4 R 5 N-SO 2 ; wherein R is H, Ci -]2 alkyl, C 3-10 cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, aryloxyalkyl, heteroarylalkyl, heteroaryloxyalkyl, or heteroarylalkoxyalkyl; and where R 4 and R 5 are each independently H, Ci.
  • Xi (where applicable), X 2 , and X 3 are N-H or N-alkylated, (e.g., N-methylated) amino acid residues.
  • the polypeptide may be a 10-mer to 15- mer and capable of binding to and activating the GLP- 1 receptor.
  • NaI naphthylalanine
  • NmA N-methylalanine
  • Aib a-aminoisobutyric acid
  • any of the peptide GLP-I analogs described herein may be modified (e.g., as described herein or as known in the art.
  • the polypeptide can be bound to a polymer to increase its molecular weight.
  • Exemplary polymers include polyethylene glycol polymers, polyamino acids, albumin, gelatin, succinyl-gelatin, (hydroxypropyl)- methacrylamide, fatty acids, polysaccharides, lipid amino acids, and dextran.
  • the polypeptide is modified by addition of albumin (e.g., human albumin), or an analog or fragment thereof, or the Fc portion of an immunoglobulin. Such an approach is described, for example, in U.S. Patent No. 7,271,149.
  • the polypeptide is modified by addition of a lipophilic substituent, as described in PCT Publication WO 98/08871.
  • the lipophilic substituent may include a partially or completely hydrogenated cyclopentanophenathrene skeleton, a straight-chain or branched alkyl group; the acyl group of a straight-chain or branched fatty acid (e.g., a group including CH 3 (CH 2 ) n CO- or HOOC(CH 2 ) m CO-, where n or m is 4 to 38); an acyl group of a straight-chain or branched alkane ⁇ , ⁇ -dicarboxylic acid;
  • the GLP-I peptide is modified by addition of a chemically reactive group such as a maleimide group, as described in U.S. Patent No. 6,593,295.
  • a chemically reactive group such as a maleimide group
  • these groups can react with available reactive functionalities on blood components to form covalent bonds and can extending the effective therapeutic in vivo half-life of the modified insulinotropic peptides.
  • a chemically reactive group a wide variety of active carboxyl groups (e.g., esters) where the hydroxyl moiety is physiologically acceptable at the levels required to modify the peptide.
  • Particular agents include N- hydroxysuccinimide (NHS), N-hydroxy-sulfosuccinimide (sulfo-NHS), maleimide-benzoyl-succinimide (MBS), gamma-maleimido-butyryloxy succinimide ester (GMBS), maleimido propionic acid (MPA) maleimido hexanoic acid (MHA), and maleimido undecanoic acid (MUA).
  • NHS N- hydroxysuccinimide
  • sulfo-NHS N-hydroxy-sulfosuccinimide
  • MBS gamma-maleimido-butyryloxy succinimide ester
  • MHA maleimido propionic acid
  • MHA maleimido hexanoic acid
  • MUA maleimido undecanoic acid
  • Primary amines are the principal targets for NHS esters. Accessible ⁇ - amine groups present on the N-termini of proteins and the ⁇ -amine of lysine react with NHS esters. An amide bond is formed when the NHS ester conjugation reaction reacts with primary amines releasing N- hydroxysuccinimide.
  • succinimide containing reactive groups are herein referred to as succinimidyl groups.
  • the functional group on the protein will be a thiol group and the chemically reactive group will be a maleimido-containing group such as gamma- maleimide-butrylamide (GMBA or MPA). Such maleimide containing groups are referred to herein as maleido groups.
  • the maleimido group is most selective for sulfhydryl groups on peptides when the pH of the reaction mixture is 6.5-7.4.
  • the rate of reaction of maleimido groups with sulfhydryls e.g., thiol groups on proteins such as serum albumin or IgG
  • sulfhydryls e.g., thiol groups on proteins such as serum albumin or IgG
  • a stable thioether linkage between the maleimido group and the sulfhydryl is formed, which cannot be cleaved under physiological conditions.
  • the compounds of the invention can feature any of polypeptides described herein, for example, any of the peptides described in Table 1 (e.g., Angiopep-1 or Angiopep-2), or a fragment or analog thereof.
  • the polypeptide may have at least 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or even 100% identity to a polypeptide described herein.
  • the polypeptide may have one or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15) substitutions relative to one of the sequences described herein. Other modifications are described in greater detail below.
  • the invention also features fragments of these polypeptides (e.g., a functional fragment).
  • the fragments are capable of efficiently being transported to or accumulating in a particular cell type (e.g., liver, eye, lung, kidney, or spleen) or are efficiently transported across the BBB.
  • Truncations of the polypeptide may be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more amino acids from either the N-terminus of the polypeptide, the C-terminus of the polypeptide, or a combination thereof.
  • Other fragments include sequences where internal portions of the polypeptide are deleted.
  • Additional polypeptides may be identified by using one of the assays or methods described herein.
  • a candidate polypeptide may be produced by conventional peptide synthesis, conjugated with paclitaxel and administered to a laboratory animal.
  • a biologically-active polypeptide conjugate may be identified, for example, based on its ability to increase survival of an animal injected with tumor cells and treated with the conjugate as compared to a control which has not been treated with a conjugate (e.g., treated with the unconjugated agent).
  • a biologically active polypeptide may be identified based on its location in the parenchyma in an in situ cerebral perfusion assay.
  • Labelled conjugates of a polypeptide can be administered to an animal, and accumulation in different organs can be measured.
  • a PATENT for example, a PATENT
  • ATTORNEY DOCKET NO. V82690WO polypeptide conjugated to a detectable label allows live in vivo visualization.
  • a detectable label e.g., a near-IR fluorescence spectroscopy label such as Cy5.5
  • a polypeptide can be administered to an animal, and the presence of the polypeptide in an organ can be detected, thus allowing determination of the rate and amount of accumulation of the polypeptide in the desired organ.
  • the polypeptide can be labelled with a radioactive isotope (e.g., ' 25 I). The polypeptide is then administered to an animal. After a period of time, the animal is sacrificed and the organs are extracted. The amount of radioisotope in each organ can then be measured using any means known in the art.
  • a labeled candidate polypeptide in a particular organ By comparing the amount of a labeled candidate polypeptide in a particular organ relative to the amount of a labeled control polypeptide, the ability of the candidate polypeptide to access and accumulate in a particular tissue can be ascertained.
  • Appropriate negative controls include any peptide or polypeptide known not to be efficiently transported into a particular cell type (e.g., a peptide related to Angiopep that does not cross the BBB, or any other peptide).
  • aprotinin analogs may be found by performing a protein BLAST (Genbank: www.ncbi.nlm.nih.gov/BLAST/) using the synthetic aprotinin sequence (or portion thereof) disclosed in International Application No.
  • PCT/CA2004/000011 Exemplary aprotinin analogs are also found under accession Nos. CAA37967 (GI:58005) and 1405218C (GL3604747). PATENT
  • the peptide vectors and peptide GLP-I agonists used in the invention may have a modified amino acid sequence.
  • the modification does not destroy significantly a desired biological activity (e.g., ability to cross the BBB or GLP-I agonist activity).
  • the modification may reduce (e.g., by at least 5%, 10%, 20%, 25%, 35%, 50%, 60%, 70%, 75%, 80%, 90%, or 95%), may have no effect, or may increase (e.g., by at least 5%, 10%, 25%, 50%, 100%, 200%, 500%, or 1000%) the biological activity of the original polypeptide.
  • the modified peptide may have or may optimize a characteristic of a polypeptide, such as in vivo stability, bioavailability, toxicity, immunological activity, immunological identity, and conjugation properties.
  • Modifications include those by natural processes, such as posttranslational processing, or by chemical modification techniques known in the art. Modifications may occur anywhere in a polypeptide including the polypeptide backbone, the amino acid side chains and the amino- or carboxy- terminus. The same type of modification may be present in the same or varying degrees at several sites in a given polypeptide, and a polypeptide may contain more than one type of modification. Polypeptides may be branched as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslational natural processes or may be made synthetically.
  • modifications include pegylation, acetylation, acylation, addition of acetomidomethyl (Acm) group, ADP-ribosylation, alkylation, amidation, biotinylation, carbamoylation, carboxyethylation, esterification, covalent attachment to flavin, covalent attachment to a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of drug, covalent attachment of a marker (e.g., fluorescent or radioactive), covalent attachment of a lipid or lipid derivative, covalent attachment of phosphatidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent crosslinks, formation of cystine, formation of pyroglutamate, formylation, gamma- PATENT
  • a modified polypeptide can also include an amino acid insertion, deletion, or substitution, either conservative or non-conservative (e.g., D-amino acids, desamino acids) in the polypeptide sequence (e.g., where such changes do not substantially alter the biological activity of the polypeptide).
  • cysteine residues can facilitate conjugation of these polypeptides by, e.g., disulfide bonding.
  • Angiopep-1 SEQ ID NO:67
  • Angiopep-2 SEQ ID NO:97
  • Angiopep-7 SEQ ID NO: 112
  • SEQ ID NOS: 71, 113, and 115 can be modified to include a single cysteine residue at the amino- terminus (SEQ ID NOS: 71, 113, and 115, respectively) or a single cysteine residue at the carboxy-terminus (SEQ ID NOS: 72, 114, and 116, respectively).
  • Amino acid substitutions can be conservative (i.e., wherein a residue is replaced by another of the same general type or group) or non-conservative (i.e., wherein a residue is replaced by an amino acid of another type).
  • a non-naturally occurring amino acid can be substituted for a naturally occurring amino acid (i.e., non-naturally occurring conservative amino acid substitution or a non-naturally occurring non-conservative amino acid substitution).
  • Polypeptides made synthetically can include substitutions of amino acids not naturally encoded by DNA (e.g., non-naturally occurring or unnatural amino acid).
  • non-naturally occurring amino acids include D- amino acids, an amino acid having an acetylaminomethyl group attached to a sulfur atom of a cysteine, a pegylated amino acid, the omega amino acids of the formula NH 2 (CH 2 ) n COOH wherein n is 2-6, neutral nonpolar amino acids, such as sarcosine, t-butyl alanine, t-butyl glycine, N-methyl isoleucine, and norleucine.
  • Phenylglycine may substitute for Tip, Tyr, or Phe; citrulline and PATENT
  • ATTORNEY DOCKET NO. V82690WO methionine sulfoxide are neutral nonpolar, cysteic acid is acidic, and ornithine is basic. Proline may be substituted with hydroxyproline and retain the conformation conferring properties.
  • Analogs may be generated by substitutional mutagenesis and retain the biological activity of the original polypeptide. Examples of substitutions identified as “conservative substitutions” are shown in Table 2. If such substitutions result in a change not desired, then other type of substitutions, denominated “exemplary substitutions” in Table 3, or as further described herein in reference to amino acid classes, are introduced and the products screened.
  • Substantial modifications in function or immunological identity are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
  • Naturally occurring residues are divided into groups based on common side chain properties:
  • Trp Tryptophan
  • Tyrosine Tyrosine
  • Phe Phenylalanine
  • Histidine His
  • polypeptides consisting of naturally occurring amino acids
  • polypeptide analogs are also encompassed by the present invention and can form the peptide vectors or GLP-I agonists used in the compounds of the invention.
  • Polypeptide analogs are commonly used in the pharmaceutical industry as non-peptide drugs with properties analogous to those of the template polypeptide.
  • the non-peptide compounds are termed "peptide mimetics" or peptidomimetics (Fauchere et al., Infect. Immun. 54:283- 287,1986 and Evans et al., J. Med. Chem. 30:1229-1239, 1987).
  • ATTORNEY DOCKET NO. V82690WO mimetics that are structurally related to therapeutically useful peptides or polypeptides may be used to produce an equivalent or enhanced therapeutic or prophylactic effect.
  • polypeptide mimetics may have significant advantages over naturally occurring polypeptides including more economical production, greater chemical stability, enhanced pharmacological properties (e.g., half-life, absorption, potency, efficiency), reduced antigenicity, and others.
  • peptide vectors described herein may efficiently cross the BBB or target particular cell types (e.g., those described herein), their effectiveness may be reduced by the presence of proteases. Likewise, the effectiveness of GLP- 1 agonists used in the invention may be similarly reduced.
  • Serum proteases have specific substrate requirements, including L-amino acids and peptide bonds for cleavage.
  • exopeptidases which represent the most prominent component of the protease activity in serum, usually act on the first peptide bond of the polypeptide and require a free N-terminus (Powell et al., Pharm. Res. 10: 1268-1273, 1993). In light of this, it is often advantageous to use modified versions of polypeptides. The modified polypeptides retain the structural characteristics of the original L-amino acid polypeptides, but advantageously are not readily susceptible to cleavage by protease and/or exopeptidases.
  • a polypeptide derivative or peptidomimetic as described herein may be all L-, all D-, or mixed D, L polypeptides.
  • the presence of an N-te ⁇ ninal or C-terminal D-amino acid increases the in vivo stability of a polypeptide because peptidases cannot utilize a D-amino acid as a substrate (Powell et al., Pharm. Res. 10:1268-1273, 1993).
  • Reverse-D polypeptides are polypeptides containing D- amino acids, arranged in a reverse sequence relative to a polypeptide containing L-amino acids.
  • the C-terminal residue of an L-amino acid polypeptide becomes N-terminal for the D-amino acid polypeptide, and so forth.
  • Reverse D-polypeptides retain the same tertiary conformation and therefore the same activity, as the L-amino acid polypeptides, but are more stable to enzymatic degradation in vitro and in vivo, and thus have greater therapeutic efficacy than the original polypeptide (Brady and Dodson, Nature 368:692-693, 1994 and Jameson et al., Nature 368:744-746, 1994).
  • constrained polypeptides comprising a consensus sequence or a substantially identical consensus sequence variation may be generated by methods well known in the art (Rizo et al., Ann. Rev. Biochem. 61:387-418, 1992).
  • constrained polypeptides may be generated by adding cysteine residues capable of forming disulfide bridges and, thereby, resulting in a cyclic polypeptide.
  • Cyclic polypeptides have no free N- or C-termini. Accordingly, they are not susceptible to proteolysis by exopeptidases, although they are, of course, susceptible to endopeptidases, which do not cleave at polypeptide termini.
  • amino acid sequences of the polypeptides with N- terminal or C-terminal D-amino acids and of the cyclic polypeptides are usually identical to the sequences of the polypeptides to which they correspond, except for the presence of N-terminal or C-terminal D-amino acid residue, or then- circular structure, respectively.
  • a cyclic derivative containing an intramolecular disulfide bond may be prepared by conventional solid phase synthesis while incorporating suitable S- protected cysteine or homocysteine residues at the positions selected for cyclization such as the amino and carboxy termini (Sah et al., J. Pharm. Pharmacol. 48:197, 1996).
  • cyclization can be performed either (1) by selective removal of the S-protecting group with a consequent on-support oxidation of the corresponding two free SH-functions, to form a S-S bonds, followed by conventional removal of the product from the support and appropriate purification procedure or (2) by removal of the polypeptide from the support along with complete side chain de- protection, followed by oxidation of the free SH-functions in highly dilute aqueous solution.
  • the cyclic derivative containing an intramolecular amide bond may be prepared by conventional solid phase synthesis while incorporating suitable amino and carboxyl side chain protected amino acid derivatives, at the position selected for cyclization.
  • the cyclic derivatives containing intramolecular -S- alkyl bonds can be prepared by conventional solid phase chemistry while incorporating an amino acid residue with a suitable amino-protected side chain, and a suitable S-protected cysteine or homocysteine residue at the position selected for cyclization.
  • Another effective approach to confer resistance to peptidases acting on the N-terminal or C-terminal residues of a polypeptide is to add chemical groups at the polypeptide termini, such that the modified polypeptide is no longer a substrate for the peptidase.
  • One such chemical modification is glycosylate on of the polypeptides at either or both termini.
  • Certain chemical modifications, in particular N-terminal glycosylation have been shown to increase the stability of polypeptides in human serum (Powell et al., Pharm. Res. 10:1268-1273, 1993).
  • Other chemical modifications which enhance serum stability include, but are not limited to, the addition of an N-terminal alkyl group, consisting of a lower alkyl of from one to twenty carbons, such as an PATENT
  • the present invention includes modified polypeptides consisting of polypeptides bearing an N-terrninal acetyl group and/or a C- terminal amide group. Also included by the present invention are other types of polypeptide derivatives containing additional chemical moieties not normally part of the polypeptide, provided that the derivative retains the desired functional activity of the polypeptide.
  • N-acyl derivatives of the amino terminal or of another free amino group examples include (1) N-acyl derivatives of the amino terminal or of another free amino group, wherein the acyl group may be an alkanoyl group (e.g., acetyl, hexanoyl, octanoyl) an aroyl group (e.g., benzoyl) or a blocking group such as F-moc (fluorenylmethyl-O-CO-); (2) esters of the carboxy terminal or of another free carboxy or hydroxyl group; (3) amide of the carboxy-terminal or of another free carboxyl group produced by reaction with ammonia or with a suitable amine; (4) phosphorylated derivatives; (5) derivatives conjugated to an antibody or other biological ligand and other types of derivatives.
  • the acyl group may be an alkanoyl group (e.g., acetyl, hexanoyl, octanoyl) an
  • polypeptides or derivatives of the polypeptides described herein having an extension, desirably the extension does not destroy the cell targeting activity of the polypeptides or its derivatives.
  • derivatives included in the present invention are dual polypeptides consisting of two of the same, or two different polypeptides, as described herein, covalently linked to one another either directly or through a spacer, such as by a short stretch of alanine residues or by a putative site for proteolysis (e.g., by cathepsin, see e.g., U.S. Patent No. 5,126,249 and European Patent No. 495 049).
  • Multimers of the polypeptides described herein consist of a polymer of molecules formed from the same or different polypeptides or derivatives thereof.
  • the present invention also encompasses polypeptide derivatives that are chimeric or fusion proteins containing a polypeptide described herein, or fragment thereof, linked at its amino- or carboxy-terminal end, or both, to an amino acid sequence of a different protein.
  • a chimeric or fusion protein may be produced by recombinant expression of a nucleic acid encoding the protein.
  • a chimeric or fusion protein may contain at least 6 amino acids shared with one of the described polypeptides which desirably results in a chimeric or fusion protein that has an equivalent or greater functional activity.
  • non-peptidyl compounds generated to replicate the backbone geometry and pharmacophore display (peptidomimetics) of the polypeptides described herein often possess attributes of greater metabolic stability, higher potency, longer duration of action, and better bioavailability.
  • Peptidomimetics compounds can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including biological libraries, spatially addressable parallel solid phase or solution phase libraries, synthetic library methods requiring deconvolution, the 'one-bead one- compound' library method, and synthetic library methods using affinity chromatography selection.
  • biological library approach is limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer, or small molecule libraries of compounds (Lam, Anticancer Drug PATENT
  • Libraries of compounds may be presented in solution (e.g., Houghten, Biotechniques 13:412-421, 1992) or on beads (Lam, Nature 354:82-84, 1991), chips (Fodor, Nature 364:555-556, 1993), bacteria or spores (U.S. Patent No. 5,223,409), plasmids (Cull et al., Proc. Natl. Acad. Sci. USA 89: 1865- 1869, 1992) or on phage (Scott and Smith, Science 249:386-390, 1990), or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product.
  • polypeptide as described herein can be isolated and purified by any number of standard methods including, but not limited to, differential solubility (e.g., precipitation), centrifugation, chromatography (e.g., affinity, ion exchange, and size exclusion), or by any other standard techniques used for the purification of peptides, peptidomimetics, or proteins.
  • differential solubility e.g., precipitation
  • centrifugation e.g., centrifugation
  • chromatography e.g., affinity, ion exchange, and size exclusion
  • the functional properties of an identified polypeptide of interest may be evaluated using any functional assay known in the art. Desirably, assays for evaluating downstream receptor function in intracellular signaling are used (e.g., cell proliferation).
  • the peptidomimetics compounds of the present invention may be obtained using the following three-phase process: (1) scanning the polypeptides described herein to identify regions of secondary structure necessary for targeting the particular cell types described herein; (2) using conformationally constrained dipeptide surrogates to refine the backbone geometry and provide organic platforms corresponding to these surrogates; and (3) using the best organic platforms to display organic pharmocophores in libraries of candidates designed to mimic the desired activity of the native PATENT
  • ATTORNEY DOCKET NO. V82690WO polypeptide In more detail the three phases are as follows. In phase 1, the lead candidate polypeptides are scanned and their structure abridged to identify the requirements for their activity. A series of polypeptide analogs of the original are synthesized. In phase 2, the best polypeptide analogs are investigated using the conformational ⁇ constrained dipeptide surrogates. Indolizidin-2-one, indolizidin-9-one and quinolizidinone amino acids (I 2 aa, I 9 aa and Qaa respectively) are used as platforms for studying backbone geometry of the best peptide candidates.
  • the compounds of the present invention also include molecules that share the structure, polarity, charge characteristics and side chain properties of the polypeptides described herein.
  • those skilled in the art can develop peptides and peptidomimetics screening assays which are useful for identifying compounds for targeting an agent to particular cell types (e.g., those described herein).
  • the assays of this invention may be developed for low- throughput, high-throughput, or ultra-high throughput screening formats.
  • Assays of the present invention include assays amenable to automation.
  • the GLP-I agonist may be bound to the vector peptide either directly (e.g., through a covalent bond such as a peptide bond) or may be bound through a linker.
  • Linkers include chemical linking agents (e.g., cleavable linkers) and peptides.
  • the linker is a chemical linking agent.
  • the GLP- 1 agonist and vector peptide may be conjugated through sulfhydryl groups, amino groups (amines), and/or carbohydrates or any appropriate reactive group.
  • Homobifunctional and heterobifunctional cross-linkers (conjugation agents) are available from many commercial sources. Regions available for cross- linking may be found on the polypeptides of the present invention.
  • the cross- linker may comprise a flexible arm, e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 carbon atoms.
  • Exemplary cross-linkers include BS3 ([Bis(sulfosuccinimidyl)suberate]; B S3 is a homobifunctional N- hydroxysuccinimide ester that targets accessible primary amines), NHS/EDC (N-hydroxysuccinimide and N-ethyl-'(dimethylaminopropyl)carbodimide; NHS/EDC allows for the conjugation of primary amine groups with carboxyl groups), sulfo-EMCS ([N-e-Maleimidocaproic acid]hydrazide; sulfo-EMCS are heterobifunctional reactive groups (maleimide and NHS-ester) that are reactive toward sulfhydryl and amino groups), hydrazide (most proteins contain exposed carbohydrates and hydrazide is a useful reagent for linking carboxyl groups to primary amines), and SATA (N-succinimidyl-S-acetylthioacetate; SATA is reactive towards
  • active carboxyl groups e.g., esters
  • Particular agents include N-hydroxysuccinimide (NHS), N-hydroxy- sulfosuccinimide (sulfo-NHS), maleimide-benzoyl-succinimide (MBS), gamma-maleimido-butyryloxy succinimide ester (GMBS), maleimido propionic PATENT
  • Primary amines are the principal targets for NHS esters. Accessible ⁇ - amine groups present on the N-termini of proteins and the ⁇ -amine of lysine react with NHS esters. An amide bond is formed when the NHS ester conjugation reaction reacts with primary amines releasing N- hydroxysuccinimide.
  • succinimide containing reactive groups are herein referred to as succinimidyl groups.
  • the functional group on the protein will be a thiol group and the chemically reactive group will be a maleimido-containing group such as gamma- maleimide-butrylamide (GMBA or MPA). Such maleimide containing groups are referred to herein as maleido groups.
  • the maleimido group is most selective for sulfhydryl groups on peptides when the pH of the reaction mixture is 6.5-7.4.
  • the rate of reaction of maleimido groups with sulfhydryls e.g., thiol groups on proteins such as serum albumin or IgG
  • sulfhydryls e.g., thiol groups on proteins such as serum albumin or IgG
  • a stable thioether linkage between the maleimido group and the sulfhydryl can be formed.
  • the linker includes at least one amino acid (e.g., a peptide of at least 2, 3, 4, 5, 6, 7, 10, 15, 20, 25, 40, or 50 amino acids).
  • the linker is a single amino acid (e.g., any naturally occurring amino acid such as Cys).
  • a glycine-rich peptide such as a peptide having the sequence [Gly-Gly-Gly-Gly-Ser] n where n is 1, 2, 3, 4, 5 or 6 is used, as described in U.S. Patent No. 7,271,149.
  • a serine-rich peptide linker is used, as described in U.S. Patent No. 5,525,491.
  • Serine rich peptide linkers include those of the formula [X-X- X-X-GIyJ y , where up to two of the X are Thr, and the remaining X are Ser, and y is 1 to 5 (e.g., Ser-Ser-Ser-Ser-Gly, where y is greater than 1).
  • the linker is a single amino acid (e.g., any amino acid, such as GIy or Cys).
  • linkers are succinic acid, Lys, GIu, and Asp, or a dipeptide such as Gly-Lys.
  • the linker is succinic acid
  • one carboxyl group thereof may form an amide bond with an amino group of the amino acid residue
  • the other carboxyl group thereof may, for example, form an amide bond with an amino group of the peptide or substituent.
  • the linker is Lys, GIu, or Asp
  • the carboxyl group thereof may form an amide bond with an amino group of the amino acid residue
  • the amino group thereof may, for example, form an amide bond with a carboxyl group of the substituent.
  • a further linker may be inserted between the ⁇ -amino group of Lys and the substituent.
  • the further linker is succinic acid which, e.g., forms an amide bond with the ⁇ - amino group of Lys and with an amino group present in the substituent.
  • the further linker is GIu or Asp (e.g., which forms an amide bond with the ⁇ -amino group of Lys and another amide bond with a carboxyl group present in the substituent), that is, the substituent is a N ⁇ -acylated lysine residue.
  • Cyclic AMP (cAMP) production from cells expressing a GLP-I receptor can be measured in the presence and in the absence of a compound, where an increase in cAMP production indicates the compound to be a GLP-I agonist.
  • a GLP-I receptor e.g., a human receptor
  • BHK cells expressing the cloned human GLP- 1 receptor (BHK-467- 12A) were grown in DMEM media with the addition of 100 IU/ml penicillin, 100 ⁇ g/ml streptomycin, 5% fetal calf serum, and 0.5 mg/mL Geneticin G-418 (Life Technologies). The cells were washed twice in phosphate buffered saline and harvested with Versene. Plasma membranes were prepared from the cells by homogenisation with an Ultraturrax in buffer 1 (20 mM HEPES-Na, 10 mM EDTA, pH 7.4). The homogenate was PATENT
  • ATTORNEY DOCKET NO. V82690WO centrifuged at 48,000*g for 15 min at 4° C.
  • the pellet was suspended by homogenization in buffer 2 (20 mM HEPES-Na, 0.1 mM EDTA, pH 7.4), then centrifuged at 48,000*g for 15 min at 4° C. The washing procedure was repeated one more time. The final pellet was suspended in buffer 2 and used immediately for assays or stored at -80° C.
  • the functional receptor assay was carried out by measuring cAMP as a response to stimulation by the insulinotropic agent, c AMP formed was quantified by the AlphaScreenTM cAMP Kit (Perkin Elmer Life Sciences). Incubations were carried out in half-area 96-well microtiter plates in a total volume of 50 ⁇ L buffer 3 (50 mM Tris-HCl, 5 mM HEPES, 10 mM MgCl 2 , pH 7.4) and with the following additions: 1 mM ATP, 1 ⁇ M GTP, 0.5 mM 3- isobutyl-1-methylxanthine (IBMX), 0.01% Tween-20, 0.1% BSA, 6 ⁇ g membrane preparation, 15 ⁇ g/ml acceptor beads, 20 ⁇ g/ml donor beads preincubated with 6 nM biotinyl-cAMP.
  • buffer 3 50 mM Tris-HCl, 5 mM HEPES, 10 mM MgCl 2 , pH 7.
  • the compounds of the invention can be used in any therapeutic application where a GLP- 1 agonist activity in the brain, or in particular tissues, is desired.
  • GLP-I agonist activity is associated with stimulation of insulin secretion (i.e., to act as an incretin hormone) and inhibition glucagon secretion, thereby contributing to limit postprandial glucose excursions.
  • GLP-I agonists can also inhibit gastrointestinal motility and secretion, thus acting as an enterogastrone and part of the "ileal brake" mechanism.
  • GLP-I also appears to be a physiological regulator of appetite and food intake. Because of these PATENT
  • GLP-I and GLP-I receptor agonists can be used for therapy of metabolic disorders, as reviewed in, e.g., Kinzig et al., J Neurosci 23:6163- 6170, 2003.
  • Such disorders include obesity, hyperglycemia, dyslipidemia, hypertriglyceridemia, syndrome X, insulin resistance, IGT, diabetic dyslipidemia, hyperlipidemia, a cardiovascular disease, and hypertension.
  • GLP-I is also has neurological effects including sedative or anti- anxiolytic effects, as described in U.S. Patent No. 5,846,937.
  • GLP-I agonists can be used in the treatment of anxiety, aggression, psychosis, seizures, panic attacks, hysteria, or sleep disorders.
  • GLP-I agonists can also be used to treat Alzheimer's disease, as GLP-I agonists have been shown to protect neurons against amyloid- ⁇ peptide and glutamate-induced apoptosis (Perry et al., Curr Alzheimer Res 2:377-85, 2005).
  • GLP-I agonists include improving learning, enhancing neuroprotection, and alleviating a symptom of a disease or disorder of the central nervous system, e.g., through modulation of neurogenesis, and e.g., Parkinson's Disease, Alzheimer's Disease, Huntington's Disease, ALS, stroke, ADD, and neuropsychiatric syndromes (U.S. Patent No. 6,969,702 and U.S. Patent Application No. 2002/0115605). Stimulation of neurogenesis using GLP-I agonists has been described, for example, in Bertilsson et al., J Neurosci Res 86:326-338, 2008.
  • Still other therapeutic uses include converting liver stem/progenitor cells into functional pancreatic cells (U.S. Patent Application Publication No. 2005/0053588); preventing beta-cell deterioration (U.S. Patent Nos. 7,259,233 and 6,569,832) and stimulation of beta-cell proliferation (U.S. Patent Application Publication No. 2003/0224983); treating obesity (U.S. Patent No. 7,211,557); suppressing appetite and inducing satiety (U.S. Patent Application Publication No. 2003/0232754); treating irritable bowel syndrome (U.S. Patent No. 6,348,447); reducing the morbidity and/or mortality associated with myocardial infarction (US Patent No. 6,747,006) and stroke (PCT Publication No. WO 00/16797); treating acute coronary syndrome characterized by an PATENT
  • ATTORNEY DOCKET NO. V82690WO absence of Q-wave myocardial infarction U.S. Patent No. 7,056,887
  • attenuating post-surgical catabolic changes U.S. Patent No. 6,006,753
  • treating hibernating myocardium or diabetic cardiomyopathy U.S. Patent No. 6,894,024
  • suppressing plasma blood levels of norepinepherine U.S. Patent No. 6,894,024
  • increasing urinary sodium excretion, decreasing urinary potassium concentration U.S. Patent No.
  • nephropathy U.S. Patent Application Publication No. 2004/0209803
  • left ventricular systolic dysfunction e.g., with abnormal left ventricular ejection fraction
  • antro-duodenal motility e.g., for the treatment or prevention of gastrointestinal disorders such as diarrhea, postoperative dumping syndrome and irritable bowel syndrome, and as premedication in endoscopic procedures
  • CIPN critical illness polyneuropathy
  • SIRS systemic inflammatory response syndrome
  • the present invention also features pharmaceutical compositions that contain a therapeutically effective amount of a compound of the invention.
  • the composition can be formulated for use in a variety of drug delivery systems.
  • One or more physiologically acceptable excipients or carriers can also be included in the composition for proper formulation.
  • Suitable formulations for use in the present invention are found in Remington 's Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, PA, 17th ed., 1985.
  • Langer Science 249:1527-1533, 1990).
  • the pharmaceutical compositions are intended for parenteral, intranasal, topical, oral, or local administration, such as by a transdermal means, for prophylactic and/or therapeutic treatment.
  • the pharmaceutical compositions can be administered parenterally (e.g., by intravenous, intramuscular, or subcutaneous injection), or by oral ingestion, or by topical application or intraarticular injection at areas affected by the vascular or cancer condition. Additional routes of administration include intravascular, intra-arterial, intratumor, intraperitoneal, intraventricular, intraepidural, as well as nasal, ophthalmic, intrascleral, intraorbital, rectal, topical, or aerosol inhalation administration.
  • compositions for parenteral administration that comprise the above mention agents dissolved or suspended in an acceptable carrier, preferably an aqueous carrier, e.g., water, buffered water, saline, PBS, and the like.
  • an acceptable carrier preferably an aqueous carrier, e.g., water, buffered water, saline, PBS, and the like.
  • the compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents, detergents and the like.
  • compositions for oral delivery which may contain inert ingredients such as binders or fillers for the formulation of a tablet, a capsule, and the like.
  • compositions for local administration which may contain inert ingredients such as solvents or emulsifiers for the formulation of a cream, an ointment, and the like.
  • compositions may be sterilized by conventional sterilization techniques, or may be sterile filtered.
  • the resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration.
  • the pH of the preparations typically will be between 3 and 11, more preferably between 5 and 9 or between 6 and 8, and most preferably between 7 and 8, such as 7 to 7.5.
  • the resulting compositions in solid form may be packaged in multiple single dose units, each containing a fixed amount of the above-mentioned agent or agents, such as in a sealed package of tablets or capsules.
  • compositions in solid form can also be packaged in a container for a flexible quantity, such as in a squeezable tube designed for a topically applicable cream or ointment.
  • the compositions containing an effective amount can be administered for prophylactic or therapeutic treatments.
  • compositions can be administered to a subject with a clinically determined predisposition or increased susceptibility to a metabolic disorder or neurological disease.
  • Compositions of the invention can be administered to the patient (e.g., a human) in an amount sufficient to delay, reduce, or preferably prevent the onset of clinical disease.
  • compositions are administered to a subject (e.g., a human) already suffering from disease (e.g., a metabolic disorder such as those described herein, or a neurological disease) in an amount sufficient to cure or at least partially arrest the symptoms of the condition and its complications.
  • disease e.g., a metabolic disorder such as those described herein, or a neurological disease
  • An amount adequate to accomplish this purpose is defined as a "therapeutically effective amount," an amount of a compound sufficient to substantially improve some symptom associated with a disease or a medical condition.
  • an agent or compound which decreases, prevents, delays, suppresses, or arrests any symptom of the disease or condition PATENT
  • ATTORNEY DOCKET NO. V82690WO would be therapeutically effective.
  • a therapeutically effective amount of an agent or compound is not required to cure a disease or condition but will provide a treatment for a disease or condition such that the onset of the disease or condition is delayed, hindered, or prevented, or the disease or condition symptoms are ameliorated, or the term of the disease or condition is changed or, for example, is less severe or recovery is accelerated in an individual.
  • Exendin-4 is typically taken twice daily at either 5 ⁇ g or 10 ⁇ g per dose for treatment of diabetes.
  • the compounds of the invention may be administered in equivalent doses of as specified for exendin-4, may be administered in higher equivalent doses (e.g., 10%, 25%, 50%, 100%, 200%, 500%, 1000% greater doses), or can be administered in lower equivalent doses (e.g., 90%, 75%, 50%, 40%, 30%, 20%, 15%, 12%, 10%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.1% of the equivalent dose).
  • equivalent doses of as specified for exendin-4 may be administered in higher equivalent doses (e.g., 10%, 25%, 50%, 100%, 200%, 500%, 1000% greater doses), or can be administered in lower equivalent doses (e.g., 90%, 75%, 50%, 40%, 30%, 20%, 15%, 12%, 10%, 8%, 7%, 6%, 5%, 4%, 3%,
  • Amounts effective for this use may depend on the severity of the disease or condition and the weight and general state of the patient, but generally range from about 0.05 ⁇ g to about 1000 ⁇ g (e.g., 0.5-100 ⁇ g) of an equivalent amount of exendin-4 the agent or agents per dose per patient.
  • Suitable regimes for initial administration and booster administrations are typified by an initial administration followed by repeated doses at one or more hourly, daily, weekly, or monthly intervals by a subsequent administration.
  • the total effective amount of an agent present in the compositions of the invention can be administered to a mammal as a single dose, either as a bolus or by infusion over a relatively short period of time, or can be administered using a fractionated treatment protocol, in which multiple doses are administered over a more prolonged period of time (e.g., a dose every 4-6, 8-12, 14-16, or 18-24 hours, or every 2-4 days, 1-2 weeks, once a month).
  • a fractionated treatment protocol in which multiple doses are administered over a more prolonged period of time (e.g., a dose every 4-6, 8-12, 14-16, or 18-24 hours, or every 2-4 days, 1-2 weeks, once a month).
  • continuous intravenous infusion sufficient to maintain therapeutically effective concentrations in the blood are contemplated.
  • the therapeutically effective amount of one or more agents present within the compositions of the invention and used in the methods of this invention applied to mammals can be determined by the PATENT
  • the dosage of the compounds of the invention can be lower than (e.g., less than or equal to about 90%, 75%, 50%, 40%, 30%, 20%, 15%, 12%, 10%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.1% of) the equivalent dose of required for a therapeutic effect of the unconjugated GLP-I agonist.
  • the agents of the invention are administered to a subject (e.g.
  • an effective amount which is an amount that produces a desirable result in a treated subject (e.g. reduction in glycemia, reduced weight gain, increased weight loss, and reduced food intake).
  • Therapeutically effective amounts can also be determined empirically by those of skill in the art.
  • the patient may also receive an agent in the range of about 0.05 to 1,000 ⁇ g equivalent dose as compared to exendin-4 per dose one or more times per week (e.g., 2, 3, 4, 5, 6, or 7 or more times per week), 0.1 to 2,500 (e.g., 2,000, 1,500, 1,000, 500, 100, 10, 1, 0.5, or 0.1) ⁇ g dose per week.
  • a patient may also receive an agent of the composition in the range of 0.1 to 3,000 ⁇ g per dose once every two or three weeks.
  • compositions of the invention comprising an effective amount can be carried out with dose levels and pattern being selected by the treating physician.
  • the dose and administration schedule can be determined and adjusted based on the severity of the disease or condition in the patient, which may be monitored throughout the course of treatment according to the methods commonly practiced by clinicians or those described herein.
  • the compounds of the present invention may be used in combination with either conventional methods of treatment or therapy or may be used separately from conventional methods of treatment or therapy.
  • compositions according to the present invention may be comprised of a combination of a compound of the present invention in association with a pharmaceutically acceptable excipient, as described herein, and another therapeutic or prophylactic agent known in the art.
  • the exemplary GLP-I conjugates, exendin-4-cysAn2 N-terminal, and Exendin-4-cysAn2 C-terminal, and Angiopep-1/Exendin 4 conjugates were made by conjugating [Lys(maleimido hexanoic acid) 39 ]exendin-4 to the sulfide in cys-An2 (SEQ ID NO:113), in An2-cys (SEQ ID NO:114), or in Angiopep-1 (SEQ ID NO:67) in Ix PBS buffer for 1 hour. This resulted in production of exendin-4/Angiopep conjugates, as shown in Figure 2.
  • a second set of exendin-4/Angiopep conjugates was made by reacting
  • Angiopep-2 having maleimido propionic acid (MPA), maleimido hexanoic acid (MHA), or maleimido undecanoic acid (MUA) bound to its N-terminus with [Cys 32 ]Exendin-4 to form a conjugate, as shown in Figure 3.
  • the common carotid was then catheterized rostrally with polyethylene tubing filled with heparin (25 U/ml) and mounted on a 26-gauge needle.
  • the syringe containing the perfusion fluid [ 125 I]-proteins or [ 125 I]-peptides in Krebs/bicarbonate buffer at pH 7.4, gassed with 95% O 2 and 5% CO 2
  • the brain was perfused for 5 min at a flow rate of 1.15 ml/min. After perfusion of radiolabeled molecules, the brain was further perfused for 60 s with Krebs buffer, to wash away excess [ 125 I]-proteins. Mice were then decapitated to terminate perfusion and the right hemisphere was isolated on ice before being subjected to capillary depletion. Aliquots of homogenates, supernatants, pellets, and perfusates were taken to measure their contents and to evaluate the apparent volume of distribution. From these experiments, brain distribution of both exendin-4/ Angiopep-
  • Obese mice (ob/ob mice) were administered the [Lys 39 -MHA]exendin- 4/Angiopep-2-Cys-NH 2 conjugate (Exen-An2).
  • PATENT [Lys 39 -MHA]exendin- 4/Angiopep-2-Cys-NH 2 conjugate
  • a 1.6 ⁇ g/kg dose of Exen-An2 is equivalent to a 1 ⁇ g/kg dose of exendin-4.
  • the body weight of each mouse was measured daily. Food intake was estimated based on the mean values for each group, and glycemia was measured one hour following treatment. After 10 days of treatment, body weight gain and food intake of mice treated at the higher doses of either exendin-4 or the conjugate are lower than the control ( Figure 5). Food intake was also reduced in the mice receiving the higher doses of either exendin-4 or the conjugate ( Figure 6) as compared to the control. Glycemia measurements showed that the lower dose of the conjugate had the same effect as the higher doses of either exendin-4 or Exen-An2 ( Figure 7). Thus, a similar effect of 1/10 the dosage on glycemia is observed using the conjugate, as compared to exendin-4.
  • Exendin-4- Angiopep-2 dimer was generated having the structure shown in Figure 8A. Briefly, the amine group in the C-terminal lysine of [Lys 39 ]Exendin-4 was conjugated to an Angiopep-2 dimer through an MHA linker at the N-terminal threonine of the first Angiopep-2 peptide. A N- Succinimidyl-S-acetylthiopropionate (SATP) linker was attached to an Angiopep-2-Cys peptide at its N-terminus. Through this cysteine, the PATENT
  • Angiopep-2-Cys peptide was conjugated to a second Angiopep-2 peptide, which had been modified to contain an MPA linker.
  • the dimer was the linked to the [Lys 39 ]Exendin-4 through an MHA linker
  • a control molecule (Exen-S4) was also generated using a scrambed form of Angiopep-2 conjugated at its N- terminal to the cysteine of [Cys 32 ]Exendin-4 through an MHA linker ( Figure 8B).
  • These conjugates were prepared as trifluoroacetate (TFA) salts.
  • Example 5 Characterization of an exendin-4-Angiopep-2 dimer conjugate Brain uptake of the exemplary GLP-I agonist, exendin-4, was measured in situ when unconjugated, conjugated to a single Angiopep-2, conjugated to a scrambled Angiopep-2 (S4), or conjugated to a dimeric form of Angiopep-2. The experiments were performed as described in Example 2 above.
  • mice were injected with a bolus containing a control, exendin-4, or the exendin-4- Angiopep-2 dimer conjugate. Mice receiving either exendin-4 or the conjugate exhibited reduced glycemia as compared to mice receiving the control ( Figure 10).
  • mice were injected with a bolus containing a control, exendin-4, or the exendin-4- Angiopep-2 dimer conjugate. Mice receiving either exendin-4 or the conjugate exhibited reduced glycemia as compared to mice receiving the control ( Figure 9).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Diabetes (AREA)
  • Endocrinology (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Cardiology (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Vascular Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Toxicology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Hospice & Palliative Care (AREA)
  • Pain & Pain Management (AREA)
  • Emergency Medicine (AREA)
  • Psychology (AREA)
  • Psychiatry (AREA)
PCT/CA2009/001476 2008-10-15 2009-10-15 Conjugates of glp-1 agonists and uses thereof WO2010043047A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
EP09820161.9A EP2346906A4 (en) 2008-10-15 2009-10-15 CONJUGATES FROM GLP-1 AGONISTS AND THEIR USE
JP2011531313A JP2012505637A (ja) 2008-10-15 2009-10-15 Glp−1アゴニストのコンジュゲート及びその使用
BRPI0920209A BRPI0920209A2 (pt) 2008-10-15 2009-10-15 conjugados de agonistas de glp-1 e usos dos mesmos
AU2009304560A AU2009304560A1 (en) 2008-10-15 2009-10-15 Conjugates of GLP-1 agonists and uses thereof
RU2011118056/10A RU2011118056A (ru) 2008-10-15 2009-10-15 Конъюгаты агонистов glp-1 и их применение
CA2740316A CA2740316A1 (en) 2008-10-15 2009-10-15 Conjugates of glp-1 agonists and uses thereof
CN2009801502163A CN102245642A (zh) 2008-10-15 2009-10-15 Glp-1激动剂的结合物及其用途
US13/124,013 US8921314B2 (en) 2008-10-15 2009-10-15 Conjugates of GLP-1 agonists and uses thereof
MX2011004017A MX2011004017A (es) 2008-10-15 2009-10-15 Conjugados de agonistas glp-1 y usos de los mismos.
ZA2011/03044A ZA201103044B (en) 2008-10-15 2011-04-21 Conjugates of glp-1 agonists and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10561808P 2008-10-15 2008-10-15
US61/105,618 2008-10-15

Publications (1)

Publication Number Publication Date
WO2010043047A1 true WO2010043047A1 (en) 2010-04-22

Family

ID=42106171

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2009/001476 WO2010043047A1 (en) 2008-10-15 2009-10-15 Conjugates of glp-1 agonists and uses thereof

Country Status (11)

Country Link
US (1) US8921314B2 (zh)
EP (1) EP2346906A4 (zh)
JP (1) JP2012505637A (zh)
CN (1) CN102245642A (zh)
AU (1) AU2009304560A1 (zh)
BR (1) BRPI0920209A2 (zh)
CA (1) CA2740316A1 (zh)
MX (1) MX2011004017A (zh)
RU (1) RU2011118056A (zh)
WO (1) WO2010043047A1 (zh)
ZA (1) ZA201103044B (zh)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011000095A1 (en) * 2009-07-02 2011-01-06 Angiochem Inc. Multimeric peptide conjugates and uses thereof
US7902156B2 (en) 2005-02-18 2011-03-08 Angiochem Inc. Aprotinin polypeptides for transporting a compound across the blood-brain barrier
WO2011153642A1 (en) * 2010-06-10 2011-12-15 Angiochem Inc. Leptin and leptin analog conjugates and fusion proteins and uses thereof
WO2012015975A3 (en) * 2010-07-28 2012-05-10 Amylin Pharmaceuticals, Inc. Glp-1 receptor agonist compounds having stabilized regions
US8487072B2 (en) 2006-10-19 2013-07-16 Angiochem Inc. Compounds for stimulating P-glycoprotein function and uses thereof
US8853353B2 (en) 2008-12-17 2014-10-07 Angiochem, Inc. Membrane type-1 matrix metalloprotein inhibitors and uses thereof
US8921314B2 (en) 2008-10-15 2014-12-30 Angiochem, Inc. Conjugates of GLP-1 agonists and uses thereof
US8969310B2 (en) 2005-07-15 2015-03-03 Angiochem Inc. Potentiation of anticancer agents
US9173891B2 (en) 2009-04-20 2015-11-03 Angiochem, Inc. Treatment of ovarian cancer using an anticancer agent conjugated to an angiopep-2 analog
US9221867B2 (en) 2003-01-06 2015-12-29 Angiochem Inc. Method for transporting a compound across the blood-brain barrier
US9365634B2 (en) 2007-05-29 2016-06-14 Angiochem Inc. Aprotinin-like polypeptides for delivering agents conjugated thereto to tissues
WO2017005734A1 (en) 2015-07-06 2017-01-12 Ucb Biopharma Sprl Tau-binding antibodies
US9687561B2 (en) 2012-08-14 2017-06-27 Angiochem Inc. Peptide-dendrimer conjugates and uses thereof
US9914754B2 (en) 2008-12-05 2018-03-13 Angiochem Inc. Conjugates of neurotensin or neurotensin analogs and uses thereof
US10251957B2 (en) 2011-06-02 2019-04-09 Hanmi Science Co., Ltd. Composition for treating diabetes comprising long-acting insulin conjugate and long-acting insulinotropic peptide conjugate
CN112079931A (zh) * 2020-08-17 2020-12-15 平顶山学院 长效glp-1类似物多肽及其降糖调脂用途
US10980892B2 (en) 2015-06-15 2021-04-20 Angiochem Inc. Methods for the treatment of leptomeningeal carcinomatosis
US11220534B2 (en) 2016-12-05 2022-01-11 Lancaster University Business Enterprises Limited Treatment of neurological diseases
WO2022178425A1 (en) * 2021-02-22 2022-08-25 Yale University Targeted bifunctional degraders
WO2023228155A1 (en) * 2022-05-27 2023-11-30 D&D Pharmatech Inc. Compositions and methods for treatment of neurological disorders

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10745456B2 (en) * 2015-04-01 2020-08-18 The Scripps Research Institute Methods and compositions related to GPCR agonist polypeptides
CN110312532A (zh) * 2016-12-19 2019-10-08 韩美药品株式会社 脑靶向长效蛋白质缀合物
EP3836951A1 (en) 2018-08-13 2021-06-23 Signablok, Inc. Peptides and compositions for targeted treatment and imaging
KR102193211B1 (ko) * 2019-11-27 2020-12-18 (주)디앤디파마텍 비오틴 모이어티가 결합된 폴리펩티드 및 이를 포함하는 경구 투여용 약학적 조성물

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006086870A1 (en) * 2005-02-18 2006-08-24 Angiochem Inc. Aprotinin polypeptides for transporting a compound across the blood-brain barrier
WO2008012629A2 (en) * 2006-07-24 2008-01-31 Biorexis Pharmaceutical Corporation Exendin fusion proteins

Family Cites Families (217)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4671958A (en) 1982-03-09 1987-06-09 Cytogen Corporation Antibody conjugates for the delivery of compounds to target sites
JPS63503086A (ja) 1986-03-13 1988-11-10 バイオテクノロジ− オ−ストラリア プロプライエタリ− リミテッド インヒビンの分析法
US4902505A (en) 1986-07-30 1990-02-20 Alkermes Chimeric peptides for neuropeptide delivery through the blood-brain barrier
US4801575A (en) 1986-07-30 1989-01-31 The Regents Of The University Of California Chimeric peptides for neuropeptide delivery through the blood-brain barrier
US4904768A (en) 1987-08-04 1990-02-27 Bristol-Myers Company Epipodophyllotoxin glucoside 4'-phosphate derivatives
GB2208511A (en) 1987-08-07 1989-04-05 Bayer Ag Variants of bovine pancreatic trypsin inhibitor produced by recombinant dna technology
US4942184A (en) 1988-03-07 1990-07-17 The United States Of America As Represented By The Department Of Health And Human Services Water soluble, antineoplastic derivatives of taxol
US5632990A (en) 1988-04-22 1997-05-27 Cancer Research Campaign Tech. Ltd. Treatment for tumors comprising conjugated antibody A5B7 and a prodrug
US5258499A (en) 1988-05-16 1993-11-02 Vestar, Inc. Liposome targeting using receptor specific ligands
US5028697A (en) 1988-08-08 1991-07-02 Eli Lilly And Company Cytotoxic antibody conjugates of hydrazide derivatized methotrexate analogs via simple organic linkers
US5169933A (en) 1988-08-15 1992-12-08 Neorx Corporation Covalently-linked complexes and methods for enhanced cytotoxicity and imaging
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5948634A (en) 1988-12-21 1999-09-07 The General Hospital Coporation Neural thread protein gene expression and detection of alzheimer's disease
DE3912638A1 (de) 1989-04-18 1990-10-31 Bayer Ag Gentechnologisch hergestellte homologe des alzheimer protease inhibitors, wirtstaemme sowie expressionsvektoren fuer ihre herstellung und ihre verwendung als arzneimittel
US5126249A (en) 1989-05-09 1992-06-30 Eli Lilly And Company Enzymatic removal of a protein amino-terminal sequence
US6020145A (en) 1989-06-30 2000-02-01 Bristol-Myers Squibb Company Methods for determining the presence of carcinoma using the antigen binding region of monoclonal antibody BR96
US6475781B1 (en) 1990-05-17 2002-11-05 Dana-Farber Cancer Institute, Inc. Trans-dominant suppressor genes for oligomeric proteins
US5714167A (en) 1992-06-15 1998-02-03 Emisphere Technologies, Inc. Active agent transport systems
GB9017008D0 (en) 1990-08-02 1990-09-19 Erba Carlo Spa Process for the enzymatic preparation of basic fibroblast growth factor
JPH07108232B2 (ja) 1990-10-09 1995-11-22 エム・ディ・リサーチ株式会社 ペプチド又は蛋白質の製造方法
US6495513B1 (en) 1991-03-11 2002-12-17 Curis, Inc. Morphogen-enhanced survival and repair of neural cells
US5955444A (en) 1991-08-09 1999-09-21 Massachusetts Institute Of Technology Method of inhibiting abnormal tau hyper phosphorylation in a cell
US5908832A (en) 1991-10-07 1999-06-01 University Of Houston - Clearlake Peptide analog
US5627270A (en) 1991-12-13 1997-05-06 Trustees Of Princeton University Glycosylated steroid derivatives for transport across biological membranes and process for making and using same
US5204354A (en) 1992-02-14 1993-04-20 Merck & Co., Inc. Substituted quinazolinones as neurotensin antagonists useful in the treatment of CNS disorders
GB9213077D0 (en) 1992-06-19 1992-08-05 Erba Carlo Spa Polymerbound taxol derivatives
JPH08504568A (ja) 1992-09-25 1996-05-21 コルバス・インターナショナル、インコーポレイテッド ウシ膵トリプシン阻害因子から誘導される因子VIIa−組織因子複合体の阻害因子
EP0599303A3 (en) 1992-11-27 1998-07-29 Takeda Chemical Industries, Ltd. Peptide conjugate
WO1994019692A1 (en) 1993-02-18 1994-09-01 The General Hospital Corporation Alzheimer's disease therapeutics
US6974578B1 (en) 1993-12-28 2005-12-13 Allergan, Inc. Method for treating secretions and glands using botulinum toxin
WO1996031531A2 (en) 1995-04-04 1996-10-10 Advanced Bioconcept, Inc. Fluorescent peptides
ZA963619B (en) 1995-05-08 1996-11-22 Scios Inc Protease inhibitor peptides
EP0827539A2 (en) 1995-05-08 1998-03-11 Scios Inc. Kunitz type protease inhibitors
AU5908296A (en) 1995-05-31 1996-12-24 Fred Hutchinson Cancer Research Center Compositions and methods for targeted delivery of effector m olecules
US5780265A (en) 1995-06-05 1998-07-14 Genentech, Inc. Kunitz type plasma kallikrein inhibitors
US5665702A (en) 1995-06-06 1997-09-09 Biomeasure Incorporated Ionic molecular conjugates of N-acylated derivatives of poly(2-amino-2-deoxy-D-glucose) and polypeptides
AU6267896A (en) 1995-06-07 1996-12-30 Imclone Systems Incorporated Antibody and antibody fragments for inhibiting the growth oftumors
HU226419B1 (en) 1996-03-11 2008-12-29 Aerovance Human bikunin
AU2409397A (en) 1996-04-24 1997-11-12 Rijksuniversiteit Te Groningen A prokaryotic protein having functional and structural homology with the uman p-glycoprotein encoded by the mdr-1 gene, nucleic acids encoding and cells expressing said protein
CA2252706A1 (en) 1996-05-01 1997-11-06 Antivirals Inc. Polypeptide conjugates for transporting substances across cell membranes
DE19636889A1 (de) 1996-09-11 1998-03-12 Felix Dr Kratz Antineoplastisch wirkende Transferrin- und Albuminkonjugate zytostatischer Verbindungen aus der Gruppe der Anthrazykline, Alkylantien, Antimetabolite und Cisplatin-Analoga und diese enthaltende Arzneimittel
AU4907897A (en) 1996-10-11 1998-05-11 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method
US6056973A (en) 1996-10-11 2000-05-02 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
US5948750A (en) 1996-10-30 1999-09-07 Merck & Co., Inc. Conjugates useful in the treatment of prostate cancer
AU6682598A (en) 1997-03-04 1998-09-22 Bio-Technology General Corporation Isolation of tissue specific peptide ligands and their use for targeting pharmaceuticals to organs
GB9705903D0 (en) 1997-03-21 1997-05-07 Elliott Gillian D VP22 Proteins and uses thereof
US6126965A (en) 1997-03-21 2000-10-03 Georgetown University School Of Medicine Liposomes containing oligonucleotides
ES2210761T3 (es) 1997-05-21 2004-07-01 The Board Of Trustees Of The Leland Stanford Junior University Composicion y procedimiento para mejorar el transporte a traves de las membranas biologicas.
US6391305B1 (en) 1997-09-10 2002-05-21 Merck & Co., Inc. Conjugates useful in the treatment of prostate cancer
US6093692A (en) 1997-09-25 2000-07-25 The University Of Southern California Method and compositions for lipidization of hydrophilic molecules
US6475481B2 (en) 1997-11-18 2002-11-05 Canji Inc Purging of stem cell products
WO1999026657A1 (en) 1997-11-25 1999-06-03 Musc Foundation For Research Development Inhibitors of nitric oxide synthase
DE69933785T2 (de) 1998-03-12 2007-10-04 Georgetown University Peptide enthaltend eine cholesterin-erkennungssequenz und deren verwendungen
US5981564A (en) 1998-07-01 1999-11-09 Universite Laval Water-soluble derivatives of paclitaxel, method for producing same and uses thereof
GB9814527D0 (en) 1998-07-03 1998-09-02 Cyclacel Ltd Delivery system
US6703381B1 (en) 1998-08-14 2004-03-09 Nobex Corporation Methods for delivery therapeutic compounds across the blood-brain barrier
US20040052814A1 (en) 1998-09-28 2004-03-18 Wenyuan Shi Fusion proteins for targeted delivery of antimicrobial peptides
US5922754A (en) 1998-10-02 1999-07-13 Abbott Laboratories Pharmaceutical compositions containing paclitaxel
AU768027B2 (en) 1999-02-24 2003-11-27 Uab Research Foundation, The Taxane derivatives for targeted therapy of cancer
ATE252601T1 (de) 1999-05-17 2003-11-15 Conjuchem Inc Lang wirkende insulinotrope peptide
GB9911683D0 (en) 1999-05-19 1999-07-21 Chiron Spa Antigenic peptides
US6180607B1 (en) 1999-08-05 2001-01-30 Christopher Davies Protein having proteinase inhibitor activity
US6713454B1 (en) 1999-09-13 2004-03-30 Nobex Corporation Prodrugs of etoposide and etoposide analogs
US6469047B1 (en) 1999-09-24 2002-10-22 Genentech, Inc. Tyrosine derivatives
US6136846A (en) 1999-10-25 2000-10-24 Supergen, Inc. Formulation for paclitaxel
DE19953696A1 (de) 1999-11-09 2001-05-10 Alexander Cherkasky Selektive proteolytische Synzyme (SPS)
GB2360453B (en) 2000-01-05 2004-09-01 Univ Cardiff Treatment of skin conditions
US20020086384A1 (en) 2000-03-14 2002-07-04 Zurit Levine Splice variants of oncogenes
US6372250B1 (en) 2000-04-25 2002-04-16 The Regents Of The University Of California Non-invasive gene targeting to the brain
AU2001285020A1 (en) 2000-08-17 2002-02-25 Synapse Technologies, Inc. P97-active agent conjugates and their methods of use
US6929919B2 (en) 2000-09-01 2005-08-16 The Governing Council Of The University Of Toronto Proteins related to schizophrenia and uses thereof
CA2355334A1 (en) 2000-10-16 2002-04-16 Procyon Biopharma Inc. Pharmaceutical preparations and methods for inhibiting tumors
JP4387102B2 (ja) 2000-10-20 2009-12-16 アボット・ラボラトリーズ 糖尿病性神経障害及び片頭痛の治療のためのチアガビンの使用
JP2004536026A (ja) 2000-11-28 2004-12-02 トランスフォーム ファーマシューティカルズ,インコーポレーティッド. パクリタキセル、その誘導体、および薬剤として許容される塩を含む医薬品製剤
PL393178A1 (pl) 2000-12-07 2011-02-14 Eli Lilly And Company Heterogenne białko fuzyjne, kompozycja farmaceutyczna do leczenia pacjentów z cukrzycą insulino-niezależną i kompozycja farmaceutyczna do leczenia pacjentów z otyłością
US7115565B2 (en) 2001-01-18 2006-10-03 Pharmacia & Upjohn Company Chemotherapeutic microemulsion compositions of paclitaxel with improved oral bioavailability
CA2441484A1 (en) 2001-03-23 2002-10-03 Napro Biotherapeutics, Inc. Molecular conjugates for use in treatment of cancer
DK1456360T3 (en) 2001-04-19 2015-08-31 Scripps Research Inst Methods and Composition for Preparation of Orthogonal TRNA-Aminoacyl-TRNA Synthetase Pairs
US20030170891A1 (en) 2001-06-06 2003-09-11 Mcswiggen James A. RNA interference mediated inhibition of epidermal growth factor receptor gene expression using short interfering nucleic acid (siNA)
US20030129186A1 (en) 2001-07-25 2003-07-10 Biomarin Pharmaceutical Inc. Compositions and methods for modulating blood-brain barrier transport
US20040161776A1 (en) 2001-10-23 2004-08-19 Maddon Paul J. PSMA formulations and uses thereof
US7192921B2 (en) 2001-11-08 2007-03-20 The Burnham Institute Peptides that home to tumor lymphatic vasculature and methods of using same
US20040058865A1 (en) 2001-11-26 2004-03-25 Danishefsky Samuel J Homing peptide multimers, their preparation and uses
BR0215184A (pt) 2001-12-20 2006-06-06 Bristol Myers Squibb Co composições farmacêuticas de derivados de taxano oralmente ativos tendo biodisponibilidade aumentada
WO2003066859A2 (en) 2002-02-07 2003-08-14 Yissum Research Development Company Of The Hebrew University Of Jerusalem Amino acid sequences capable of facilitating penetration across a biological barrier
DE60329089D1 (de) 2002-03-29 2009-10-15 Creagene Inc Zytoplasmatische transduktionspeptide und deren verwendungen
US6881829B2 (en) 2002-04-26 2005-04-19 Chimeracom, L.L.C. Chimeric hybrid analgesics
AU2003237314B2 (en) 2002-05-29 2010-03-04 Biomarin Pharmaceutical Inc. Targeted therapeutic proteins
JP2005537244A (ja) 2002-06-28 2005-12-08 ナステック・ファーマシューティカル・カンパニー・インコーポレーテッド 療法化合物の粘膜搬送を増進させるための、上皮接合部接着分子の生理機能を調節する組成物および方法
JP2006507841A (ja) 2002-11-14 2006-03-09 ダーマコン, インコーポレイテッド 機能的siRNAおよび超機能的siRNA
US7829694B2 (en) 2002-11-26 2010-11-09 Medtronic, Inc. Treatment of neurodegenerative disease through intracranial delivery of siRNA
US7319090B2 (en) 2002-11-27 2008-01-15 University Of North Carolina At Chapel Hill Methods of treating cerebral ischemia
US7388079B2 (en) 2002-11-27 2008-06-17 The Regents Of The University Of California Delivery of pharmaceutical agents via the human insulin receptor
US20040102369A1 (en) 2002-11-27 2004-05-27 The Regents Of The University Of California Transport of basic fibroblast growth factor across the blood brain barrier
CN1747747B (zh) 2003-01-06 2012-09-05 安吉奥开米公司 作为穿过血脑屏障的载体的抑酶肽及类似物
US20040162284A1 (en) 2003-02-19 2004-08-19 Harris Herbert W. Method of lowering body temperature with (S) tofisopam
JP4848267B2 (ja) 2003-04-08 2011-12-28 バイオテンプト、ビー.ヴイ. Hcg断片を含む粘膜及び経口投与のための組成物
CN104383542B (zh) 2003-04-08 2017-09-26 普罗热尼奇制药公司 包含甲基纳曲酮的药物配方
EP1466924B9 (en) 2003-04-11 2013-06-19 Institut Pasteur Synthetic peptide vaccines for HIV: the CBD epitope as an effective immunogen to elicit broadly neutralizing antibodies against HIV
EP2669377A3 (en) 2003-04-17 2015-10-14 Alnylam Pharmaceuticals Inc. Modified iRNA agents
US7374762B2 (en) 2003-05-14 2008-05-20 Immunogen, Inc. Drug conjugate composition
AU2003286870A1 (en) 2003-06-05 2005-01-04 Salk Institute For Biological Studies Targeting polypeptides to the central nervous system
US20050026823A1 (en) 2003-06-20 2005-02-03 Biomarin Pharmaceutical Inc. Use of the chaperone receptor-associated protein (RAP) for the delivery of therapeutic compounds to the brain and other tissues
CA2525236C (en) 2003-06-20 2015-03-24 Biomarin Pharmaceutical Inc. Delivery of therapeutic compounds to the brain and other tissues
JP4806258B2 (ja) 2003-08-08 2011-11-02 株式会社ACTGen 脳移行活性を有するポリペプチド、およびその利用
JP2007512001A (ja) 2003-08-28 2007-05-17 バイオレクシス ファーマシューティカル コーポレイション Epoミメティックペプチドおよび融合タンパク質
US20050058702A1 (en) 2003-09-17 2005-03-17 Ben-Sasson Shmuel A. Compositions capable of facilitating penetration across a biological barrier
US20050208095A1 (en) 2003-11-20 2005-09-22 Angiotech International Ag Polymer compositions and methods for their use
US7208174B2 (en) 2003-12-18 2007-04-24 Hoffmann-La Roche Inc. Liposome compositions
CN101035756A (zh) 2004-06-17 2007-09-12 南卡罗来纳州医科大学研究发展基金会 非天然氨基酸
MX2007000728A (es) * 2004-07-21 2007-03-15 Ambrx Inc Polipeptidos biosinteticos que utilizan amino acidos no naturalmente codificados.
JP2008507292A (ja) 2004-07-26 2008-03-13 アステリオン・リミテッド リンカー
CA2580189C (en) 2004-09-29 2013-05-21 Children's Memorial Hospital Sirna-mediated gene silencing of alpha synuclein
US9002652B1 (en) 2005-01-27 2015-04-07 Institute For Systems Biology Methods for identifying and using organ-specific proteins in blood
MX2007009960A (es) 2005-02-18 2008-03-11 Abraxis Bioscience Inc Combinaciones y metodos de administracion de agentes terapeuticos y terapia de combinacion.
US20090016959A1 (en) 2005-02-18 2009-01-15 Richard Beliveau Delivery of antibodies to the central nervous system
EP1877099B1 (en) 2005-04-06 2012-09-19 Genzyme Corporation Therapeutic conjugates comprising a lysosomal enzyme, polysialic acid and a targeting moiety
CA2616906A1 (en) 2005-05-12 2006-11-23 Tapestry Pharmaceuticals, Inc. Molecular constructs suitable for targeted conjugates
WO2006138343A2 (en) 2005-06-14 2006-12-28 Raptor Pharmaceutical Inc. Compositions comprising receptor - associated protein (rap) variants specific for lrp2 and uses thereof
WO2007009229A1 (en) 2005-07-15 2007-01-25 Angiochem Inc. Use of aprotinin polypeptides as carriers in pharmaceutical conjugates
MX2008000994A (es) 2005-07-19 2008-03-19 Univ Illinois Agentes de transporte para cruzar la barrera hematoencefalica y hacia el interior de las celulas de cancer cerebral, y metodos para el uso de los mismos.
GB0517092D0 (en) 2005-08-19 2005-09-28 Novartis Ag New compositions containing taxane derivatives
JP2009507852A (ja) 2005-09-08 2009-02-26 エムディーアールエヌエー,インコーポレイテッド リボ核酸の細胞への送達用医薬組成物
JP2009508494A (ja) 2005-09-16 2009-03-05 ラプトール ファーマシューティカル インコーポレイテッド Crを含むタンパク質に対して特異的な受容体−結合タンパク質(rap)変異体を含む組成物およびその使用
US8142781B2 (en) 2005-10-07 2012-03-27 Armagen Technologies, Inc. Fusion proteins for blood-brain barrier delivery
CA2627585A1 (en) 2005-11-01 2007-05-10 Alnylam Pharmaceuticals, Inc. Rnai inhibition of influenza virus replication
US8039432B2 (en) * 2005-11-09 2011-10-18 Conjuchem, Llc Method of treatment of diabetes and/or obesity with reduced nausea side effect
CA2633224A1 (en) 2005-12-16 2007-06-21 Argolyn Bioscience, Inc. Non-natural amino acids and neurotensin analogues thereof
US8290433B2 (en) 2007-11-14 2012-10-16 Blaze Mobile, Inc. Method and system for securing transactions made through a mobile communication device
AR054215A1 (es) 2006-01-20 2007-06-13 Eriochem Sa Una formulacion farmaceutica de un taxano, una composicion solida de un taxano liofilizado a partir de una solucion de acido acetico, un procedimiento para la preparacion de dicha composicion solida de un taxano, una composicion solubilizante de un taxano liofilizado, y un conjunto de elementos (kit
EP1996937A4 (en) 2006-03-06 2009-04-08 Amunix Inc GENETIC PACKS AND USES THEREOF
US7477504B1 (en) 2006-03-13 2009-01-13 C.P.D. Technologies, Inc. Versatile stun glove
ES2338179T3 (es) 2006-03-30 2010-05-04 Glaxo Group Limited Anticuerpos contra el peptido beta-amiloide.
JP2009543806A (ja) 2006-07-13 2009-12-10 バーナム インスティテュート フォー メディカル リサーチ gC1qR/p32を標的化するための方法および組成物
WO2008036682A2 (en) 2006-09-18 2008-03-27 Raptor Pharmaceutical Inc. Treatment of liver disorders by administration of receptor-associated protein (rap)-conjugates
CA2666841A1 (en) 2006-10-19 2008-04-24 Angiochem, Inc. Compounds for stimulating p-glycoprotein function and uses thereof
WO2008069876A2 (en) 2006-10-27 2008-06-12 University Of Kentucky Research Foundation Amidated dopamine neuron stimulating peptides for cns dopaminergic upregulation
US9399675B2 (en) 2007-02-28 2016-07-26 The Board Of Regents Of The University Of Oklahoma Methods and compositions for diagnosing Alzheimer's disease and age-related macular degeneration
KR101801454B1 (ko) 2007-03-21 2017-11-24 랩터 파마슈티컬스 인코포레이티드 환형의 수용체-연관된 단백질(rap) 펩티드
AU2008255556B2 (en) * 2007-05-29 2014-08-07 Angiochem, Inc. Aprotinin-like polypeptides for delivering agents conjugated thereto to tissues
US8802631B2 (en) 2007-09-17 2014-08-12 Ludwig Institute For Cancer Research Ltd. Peptides and methods for the treatment of gliomas and other cancers
CN104672311A (zh) 2007-10-02 2015-06-03 玛瑞纳生物技术有限公司 用于递送核酸的脂肽
WO2009070597A2 (en) 2007-11-26 2009-06-04 Armagen Technologies, Inc. Fusion proteins for delivery of gdnf to the cns
WO2009079790A1 (en) 2007-12-20 2009-07-02 Angiochem Inc. Polypeptide-nucleic acid conjugates and uses thereof
US8193182B2 (en) 2008-01-04 2012-06-05 Intellikine, Inc. Substituted isoquinolin-1(2H)-ones, and methods of use thereof
AU2009215426B2 (en) 2008-02-21 2015-06-11 Burnham Institute For Medical Research Methods and compositions related to peptides and proteins with C-terminal elements
US8710013B2 (en) 2008-04-18 2014-04-29 Angiochem Inc. Pharmaceutical compositions of paclitaxel, paclitaxel analogs or paclitaxel conjugates and related methods of preparation and use
WO2010006239A2 (en) 2008-07-10 2010-01-14 The Board Of Trustees Of The University Of Illinois Regulation of apoptosis by neural specific splice variants of ig20
BRPI0920121A2 (pt) 2008-10-15 2019-09-24 Angiochem Inc conjugados de etoposida e doxorubicina para liberação de fármaco
WO2010043047A1 (en) 2008-10-15 2010-04-22 Angiochem Inc. Conjugates of glp-1 agonists and uses thereof
MX2011005963A (es) 2008-12-05 2011-09-01 Angiochem Inc Conjugados de neurotensina o analogos de neurotensina y sus usos.
BRPI0922691A2 (pt) 2008-12-05 2018-11-06 Angiochem Inc conjugados de leptina e análogo de leptina e usos dos mesmos
CA2745499A1 (en) * 2008-12-05 2010-06-10 Angiochem Inc. Peptide therapeutic conjugates and uses thereof
CN102300987A (zh) 2008-12-17 2011-12-28 安吉奥开米公司 膜型-1基质金属蛋白抑制剂及其用途
US20110318322A1 (en) 2009-01-12 2011-12-29 Nektar Therapeutics Conjugates of a Lysosomal Enzyme Moiety and a Water Soluble Polymer
EP2421562B1 (en) 2009-04-20 2019-03-13 Angiochem Inc. Treatment of ovarian cancer using an anticancer agent conjugated to an angiopep-2 analog
US20100284921A1 (en) 2009-05-08 2010-11-11 Temple University - Of The Commonwealth System Of Higher Education Targeted nanoparticles for intracellular cancer therapy
CN102753187A (zh) 2009-06-03 2012-10-24 戴瑟纳制药公司 肽-dicer底物试剂及其特异性抑制基因表达的方法
CN103819564A (zh) 2009-06-11 2014-05-28 安吉奥开米公司 用于将gdnf和bdnf递送至中枢神经系统的融合蛋白
CN102596993A (zh) 2009-07-02 2012-07-18 安吉奥开米公司 多聚体肽结合物以及其应用
BR112012000908A2 (pt) 2009-07-14 2019-09-24 Mayo Found Medical Education & Res liberação de agentes ativos através da barreira hematoencefálica mediada por peptídeo em associação não covalente
MX2012004247A (es) 2009-10-06 2012-06-25 Angiochem Inc Composiciones y metodos para el transporte de agentes terapeuticos.
ES2655737T3 (es) 2009-11-13 2018-02-21 Daiichi Sankyo Europe Gmbh Materiales y métodos para tratar o prevenir las enfermedades asociadas a HER-3
US8530429B2 (en) 2009-11-24 2013-09-10 Arch Cancer Therapeutics, Inc. Brain tumor targeting peptides and methods
US9095541B2 (en) 2009-11-24 2015-08-04 Arch Cancer Therapeutics, Inc. Brain tumor targeting peptides and methods
EP2333074A1 (en) 2009-12-14 2011-06-15 Robert Steinfeld Substances and methods for the treatment of lysosmal storage diseases
US9180105B2 (en) 2010-03-08 2015-11-10 Sloan-Kettering Institute For Cancer Research CDC7 kinase inhibitors and uses thereof
EP2550285B1 (en) 2010-03-22 2017-07-19 President and Fellows of Harvard College Trioxacarcins and uses thereof
WO2011119608A1 (en) 2010-03-26 2011-09-29 University Of Tennessee Research Foundation Peptides that specifically target amyloid deposits
CA2796215C (en) 2010-04-14 2021-05-18 National Research Council Of Canada Compositions and methods for brain delivery of analgesic peptides
WO2011153642A1 (en) 2010-06-10 2011-12-15 Angiochem Inc. Leptin and leptin analog conjugates and fusion proteins and uses thereof
CN103080125A (zh) 2010-07-02 2013-05-01 安吉奥开米公司 用于治疗性结合物的短且含d氨基酸的多肽及其使用
US8218280B2 (en) 2010-07-08 2012-07-10 Schneider Electric USA, Inc. Secondary thermal sensor for primary conductors
JP2013540694A (ja) 2010-08-06 2013-11-07 ウー3・フアルマ・ゲー・エム・ベー・ハー Her3結合剤の前立腺治療における使用
CN102406949B (zh) 2010-09-21 2013-05-29 复旦大学 一种靶向示踪的多模式诊断纳米影像药物
WO2012037687A1 (en) 2010-09-23 2012-03-29 Angiochem Inc. Therapeutic polypeptides and uses thereof
EP2637669A4 (en) 2010-11-10 2014-04-02 Infinity Pharmaceuticals Inc Heterocyclic compounds and their use
WO2012068531A2 (en) 2010-11-18 2012-05-24 The General Hospital Corporation Novel compositions and uses of anti-hypertension agents for cancer therapy
CN102552928A (zh) 2010-12-19 2012-07-11 复旦大学 一种治疗脑部肿瘤的双级靶向递药系统及其制备方法
DK2663309T3 (en) 2011-01-10 2017-06-19 Infinity Pharmaceuticals Inc METHODS FOR PRODUCING ISOQUINOLINONES AND SOLID FORMS OF ISOQUINOLINONES
CN102614105A (zh) 2011-01-28 2012-08-01 复旦大学 一种脑靶向载两性霉素b聚合物胶束给药系统
EP2680820B1 (en) 2011-03-01 2022-11-02 2-BBB Medicines B.V. Advanced active liposomal loading of poorly water-soluble substances
CA2831392C (en) 2011-03-28 2020-04-28 Massachusetts Institute Of Technology Conjugated lipomers and uses thereof
EP2952210A3 (en) 2011-04-07 2016-03-16 Emory University Compositions comprising saccharide binding moieties and methods for targeted therapy
CN103596951A (zh) 2011-04-08 2014-02-19 阿弗拉克西斯控股股份有限公司 用于治疗神经系统障碍和癌症的8-乙基-6-(芳基)吡啶并[2,3-d]嘧啶-7(8h)-酮
EP4186501A1 (en) 2011-04-27 2023-05-31 Yale University, Inc. Drug therapy to inhibit chemotherapy-induced adverse effects and related pharmaceutical compositions, diagnostics, screening techniques and kits
AU2012251971B2 (en) 2011-05-09 2015-04-30 Institut Quimic De Sarria Cets Fundacio Privada Polymeric nanoparticles for drug delivery
WO2013004716A1 (en) 2011-07-04 2013-01-10 Universität Basel Peptide beads
EP2734520B1 (en) 2011-07-19 2016-09-14 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
CN103946226A (zh) 2011-07-19 2014-07-23 无限药品股份有限公司 杂环化合物及其应用
SG2014009492A (en) 2011-08-11 2014-09-26 Intellikine Llc Kinase inhibitor polymorphs
SG11201400310WA (en) 2011-08-29 2014-06-27 Infinity Pharmaceuticals Inc Heterocyclic compounds and uses thereof
US9630979B2 (en) 2011-09-29 2017-04-25 Infinity Pharmaceuticals, Inc. Inhibitors of monoacylglycerol lipase and methods of their use
WO2013056096A1 (en) 2011-10-13 2013-04-18 Angiochem Inc. Polypeptide-opioid conjugates and uses thereof
JP6151707B2 (ja) 2011-10-27 2017-06-21 マサチューセッツ インスティテュート オブ テクノロジー 薬物内包微小球の形成が可能なn−末端で官能基化されたアミノ酸誘導体
US9403820B2 (en) 2011-11-11 2016-08-02 Intellikine Llc Kinase inhibitor polymorphs
EP2785838A4 (en) 2011-12-01 2015-07-01 Angiochem Inc VECTORIZED LYSOSOMAL ENZYME COMPOUNDS
US20140335163A1 (en) 2011-12-01 2014-11-13 Angiochem Inc. Targeted iduronate-2-sulfatase compounds
SG10201601349XA (en) 2011-12-13 2016-03-30 Engeneic Molecular Delivery Pty Ltd Bacterially derived, intact minicells for delivery of therapeutic agents to brain tumors
AU2012351967A1 (en) 2011-12-16 2014-07-17 Massachusetts Institute Of Technology Alpha-aminoamidine polymers and uses thereof
WO2013120107A1 (en) 2012-02-09 2013-08-15 University Of Rochester Methods and compositions for treating a subject to inhibit hearing loss
US20150037281A1 (en) 2012-03-02 2015-02-05 Icahn School Of Medicine At Mount Sinai Variants of prothymosin alpha and methods of using same
WO2013151774A1 (en) 2012-04-04 2013-10-10 Halozyme, Inc. Combination therapy with an anti - hyaluronan agent and a tumor - targeted taxane
US9745334B2 (en) 2012-04-26 2017-08-29 Yale University Cytotoxic-drug delivering molecules targeting HIV (CDM-Hs), cytotoxic activity against the human immunodeficiency virus and methods of use
US20150147310A1 (en) 2012-06-15 2015-05-28 Angiochem Inc. Targeted enzyme compounds and uses thereof
CA2884870C (en) 2012-08-13 2022-03-29 Massachusetts Institute Of Technology Amine-containing lipidoids and uses thereof
AU2013302270A1 (en) 2012-08-14 2015-03-26 Angiochem Inc. Peptide-dendrimer conjugates and uses thereof
JP2015526435A (ja) 2012-08-14 2015-09-10 アンジオケム インコーポレーテッド 抗体成分と血液脳関門を通過するポリペプチドと細胞毒とを含むコンジュゲート
WO2014028777A2 (en) 2012-08-16 2014-02-20 Ipierian, Inc. Methods of treating a tauopathy
US20140120083A1 (en) 2012-11-01 2014-05-01 Infinity Pharmaceuticals, Inc. Treatment of cancers using pi3 kinase isoform modulators
PT2914296T (pt) 2012-11-01 2018-10-30 Infinity Pharmaceuticals Inc Tratamento de cancros utilizando moduladores de isoformas de pi3-quinase
CN116063483A (zh) 2012-11-02 2023-05-05 美国比奥维拉迪维股份有限公司 抗补体C1s抗体和其用途
ES2855169T3 (es) 2012-11-12 2021-09-23 Angiochem Inc Conjugados de polipéptido-anticuerpo derivados de aprotinina
GB201220474D0 (en) 2012-11-14 2012-12-26 Sagetis Biotech Sl Polypeptides
US9775915B2 (en) 2012-11-26 2017-10-03 President And Fellows Of Harvard College Trioxacarcins, trioxacarcin-antibody conjugates, and uses thereof
US20160002626A1 (en) 2012-11-28 2016-01-07 Youwen Zhou Tox inhibition for the treatment of cancer
CN104955946A (zh) 2012-11-30 2015-09-30 安吉奥开米公司 靶向的艾杜糖醛酸-2-硫酸酯酶化合物

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006086870A1 (en) * 2005-02-18 2006-08-24 Angiochem Inc. Aprotinin polypeptides for transporting a compound across the blood-brain barrier
WO2008012629A2 (en) * 2006-07-24 2008-01-31 Biorexis Pharmaceutical Corporation Exendin fusion proteins

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DEMEULE M. ET AL.: "Identification and design of peptides as a new drug delivery system for the brain.", THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, vol. 324, no. 3, 21 December 2007 (2007-12-21), pages 1064 - 1072, XP008095811 *
HARKAVYI, A. ET AL.: "Glucagon-like peptide 1 receptor stimulation reverses key deficits in distinct rodent models of Parkinson's disease.", JOURNAL OF NEUROINFLAMMATION, vol. 5, 21 May 2008 (2008-05-21), pages 19, XP021037929 *
See also references of EP2346906A4 *

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9221867B2 (en) 2003-01-06 2015-12-29 Angiochem Inc. Method for transporting a compound across the blood-brain barrier
US7902156B2 (en) 2005-02-18 2011-03-08 Angiochem Inc. Aprotinin polypeptides for transporting a compound across the blood-brain barrier
US9713646B2 (en) 2005-07-15 2017-07-25 Angiochem Inc. Potentiation of anticancer agents
US8969310B2 (en) 2005-07-15 2015-03-03 Angiochem Inc. Potentiation of anticancer agents
US8487072B2 (en) 2006-10-19 2013-07-16 Angiochem Inc. Compounds for stimulating P-glycoprotein function and uses thereof
US9365634B2 (en) 2007-05-29 2016-06-14 Angiochem Inc. Aprotinin-like polypeptides for delivering agents conjugated thereto to tissues
US8921314B2 (en) 2008-10-15 2014-12-30 Angiochem, Inc. Conjugates of GLP-1 agonists and uses thereof
US9914754B2 (en) 2008-12-05 2018-03-13 Angiochem Inc. Conjugates of neurotensin or neurotensin analogs and uses thereof
US8853353B2 (en) 2008-12-17 2014-10-07 Angiochem, Inc. Membrane type-1 matrix metalloprotein inhibitors and uses thereof
US9173891B2 (en) 2009-04-20 2015-11-03 Angiochem, Inc. Treatment of ovarian cancer using an anticancer agent conjugated to an angiopep-2 analog
WO2011000095A1 (en) * 2009-07-02 2011-01-06 Angiochem Inc. Multimeric peptide conjugates and uses thereof
US9161988B2 (en) 2009-07-02 2015-10-20 Angiochem Inc. Multimeric peptide conjugates and uses thereof
EP2448965A4 (en) * 2009-07-02 2015-02-11 Angiochem Inc MULTIMEPEPTID CONJUGATES AND ITS USES
EP2448965A1 (en) * 2009-07-02 2012-05-09 Angiochem Inc. Multimeric peptide conjugates and uses thereof
WO2011153642A1 (en) * 2010-06-10 2011-12-15 Angiochem Inc. Leptin and leptin analog conjugates and fusion proteins and uses thereof
US9217022B2 (en) 2010-07-28 2015-12-22 Astrazeneca Pharmaceuticals Lp GLP-1 receptor agonist compounds having stabilized regions
CN103200956A (zh) * 2010-07-28 2013-07-10 安米林药品有限责任公司 具有稳定区域的glp-1受体激动剂化合物
WO2012015975A3 (en) * 2010-07-28 2012-05-10 Amylin Pharmaceuticals, Inc. Glp-1 receptor agonist compounds having stabilized regions
US10251957B2 (en) 2011-06-02 2019-04-09 Hanmi Science Co., Ltd. Composition for treating diabetes comprising long-acting insulin conjugate and long-acting insulinotropic peptide conjugate
US9687561B2 (en) 2012-08-14 2017-06-27 Angiochem Inc. Peptide-dendrimer conjugates and uses thereof
US10980892B2 (en) 2015-06-15 2021-04-20 Angiochem Inc. Methods for the treatment of leptomeningeal carcinomatosis
WO2017005734A1 (en) 2015-07-06 2017-01-12 Ucb Biopharma Sprl Tau-binding antibodies
US11220534B2 (en) 2016-12-05 2022-01-11 Lancaster University Business Enterprises Limited Treatment of neurological diseases
US11851468B2 (en) 2016-12-05 2023-12-26 University Of Lancaster Treatment of neurological diseases
CN112079931A (zh) * 2020-08-17 2020-12-15 平顶山学院 长效glp-1类似物多肽及其降糖调脂用途
CN112079931B (zh) * 2020-08-17 2021-12-31 平顶山学院 长效glp-1类似物多肽及其降糖调脂用途
WO2022178425A1 (en) * 2021-02-22 2022-08-25 Yale University Targeted bifunctional degraders
WO2023228155A1 (en) * 2022-05-27 2023-11-30 D&D Pharmatech Inc. Compositions and methods for treatment of neurological disorders

Also Published As

Publication number Publication date
CN102245642A (zh) 2011-11-16
EP2346906A1 (en) 2011-07-27
ZA201103044B (en) 2012-07-25
JP2012505637A (ja) 2012-03-08
CA2740316A1 (en) 2010-04-22
US8921314B2 (en) 2014-12-30
BRPI0920209A2 (pt) 2015-12-22
RU2011118056A (ru) 2012-11-27
AU2009304560A1 (en) 2010-04-22
US20120015876A1 (en) 2012-01-19
MX2011004017A (es) 2011-06-24
EP2346906A4 (en) 2013-04-24

Similar Documents

Publication Publication Date Title
US8921314B2 (en) Conjugates of GLP-1 agonists and uses thereof
US9914754B2 (en) Conjugates of neurotensin or neurotensin analogs and uses thereof
US20110288009A1 (en) Leptin and leptin analog conjugates and uses thereof
CN108135981B (zh) 胰高血糖素受体激动剂
US9161988B2 (en) Multimeric peptide conjugates and uses thereof
JP2020007338A (ja) グルカゴン及びglp−1共アゴニスト化合物
US20160263235A1 (en) Peptide therapeutic conjugates and uses thereof
US20120196803A1 (en) Fusion proteins for delivery of gdnf and bdnf to the central nervous system
JP2017141232A (ja) 作用持続時間が増した改変ポリペプチド
WO2011153642A1 (en) Leptin and leptin analog conjugates and fusion proteins and uses thereof

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200980150216.3

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09820161

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2011531313

Country of ref document: JP

Ref document number: 2740316

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/A/2011/004017

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2009304560

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 3183/CHENP/2011

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2009820161

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2011118056

Country of ref document: RU

ENP Entry into the national phase

Ref document number: 2009304560

Country of ref document: AU

Date of ref document: 20091015

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 13124013

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0920209

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20110413